US20140128581A1 - Disubstituted amino acids and methods of preparation and use thereof - Google Patents

Disubstituted amino acids and methods of preparation and use thereof Download PDF

Info

Publication number
US20140128581A1
US20140128581A1 US14/070,306 US201314070306A US2014128581A1 US 20140128581 A1 US20140128581 A1 US 20140128581A1 US 201314070306 A US201314070306 A US 201314070306A US 2014128581 A1 US2014128581 A1 US 2014128581A1
Authority
US
United States
Prior art keywords
formula
crystalline
salt
compound
fmoc
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US14/070,306
Other versions
US9604919B2 (en
Inventor
Krzysztof Darlak
Noriyuki Kawahata
Sameer Ahmed Athamneh
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Aileron Therapeutics Inc
Original Assignee
Aileron Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Aileron Therapeutics Inc filed Critical Aileron Therapeutics Inc
Priority to US14/070,306 priority Critical patent/US9604919B2/en
Publication of US20140128581A1 publication Critical patent/US20140128581A1/en
Assigned to AILERON THERAPEUTICS, INC. reassignment AILERON THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DARLAK, KRZYSZTOF, ATHAMNEH, SAMEER AHMED, KAWAHATA, NORIYUKI
Priority to US15/278,824 priority patent/US9845287B2/en
Application granted granted Critical
Publication of US9604919B2 publication Critical patent/US9604919B2/en
Priority to US15/794,355 priority patent/US10669230B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/32Esters of carbamic acids having oxygen atoms of carbamate groups bound to carbon atoms of rings other than six-membered aromatic rings
    • C07C271/34Esters of carbamic acids having oxygen atoms of carbamate groups bound to carbon atoms of rings other than six-membered aromatic rings with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/10Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C271/22Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms to carbon atoms of hydrocarbon radicals substituted by carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C269/00Preparation of derivatives of carbamic acid, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C269/08Separation; Purification; Stabilisation; Use of additives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/006General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length of peptides containing derivatised side chain amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/02Ortho- or ortho- and peri-condensed systems
    • C07C2603/04Ortho- or ortho- and peri-condensed systems containing three rings
    • C07C2603/06Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members
    • C07C2603/10Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members containing five-membered rings
    • C07C2603/12Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members containing five-membered rings only one five-membered ring
    • C07C2603/18Fluorenes; Hydrogenated fluorenes
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • alkene ⁇ , ⁇ -Disubstituted amino acids bearing a terminal alkene on one of their side chains and their salts are useful for making cross-linked macrocyclic peptides.
  • International Application No. PCT/US2004/038403 (“the '403 application”) discloses incorporating into a peptide two ⁇ , ⁇ -disubstituted amino acids that each contain a side-chain bearing a terminal alkene, and cross-linking the terminal alkene groups to form a cross-linked (“stapled”) macrocyclic peptide.
  • the cross-link can, for example, stabilize a secondary structure (e.g., an ⁇ -helix) present in the stapled macrocyclic peptide.
  • Alkene ⁇ , ⁇ -disubstituted amino acids are thus important and useful building blocks for forming stitched and stapled polypeptides and their precursors.
  • the use of alkene ⁇ , ⁇ -disubstituted amino acids has been limited by an inability to provide these important molecules in crystalline form.
  • commercially available preparations of alkene ⁇ , ⁇ -disubstituted amino acids are typically sold as pre-made solutions.
  • the pre-made solutions limit the amount of ⁇ , ⁇ -disubstituted amino acid that can be shipped per unit volume, limit the chemical reactions that are available to be run with the alkene ⁇ , ⁇ -disubstituted amino acids, subject the alkene ⁇ , ⁇ -disubstituted amino acids to an enhanced degradation rate, and are environmentally unfriendly.
  • crystalline alkene ⁇ , ⁇ -disubstituted amino acids and their crystalline salts and processes for producing and using these crystalline amino acids.
  • substituting one or more hydrogen atoms of an alkene ⁇ , ⁇ -disubstituted amino acid with deuterium or a halogen atom can change one or more of the amino acid's properties. For example dipole moment, hydrophobicity, hydrophilicity, steric bulk, or reactivity of an alkene ⁇ , ⁇ -disubstituted amino acid can be changed by substituting one or more hydrogen atoms thereon with one or more deuterium or halogen atoms.
  • inventive embodiments provided in this Summary of the Invention are meant to be illustrative only and to provide an overview of selected inventive embodiments disclosed herein.
  • the Summary of the Invention being illustrative and selective, does not limit the scope of any claim, does not provide the entire scope of inventive embodiments disclosed or contemplated herein, and should not be construed as limiting or constraining the scope of this disclosure or any claimed inventive embodiment.
  • R 1 is C 1 -C 3 alkyl, C 1 -C 3 deuteroalkyl, or C 1 -C 3 haloalkyl; * is a stereocenter; n is an integer from 1 to 20; R 2 is —H or a nitrogen protecting group; and R 3 is —H or a protecting or activating group.
  • R 1 is C 1 -C 3 alkyl, C 1 -C 3 deuteroalkyl, or C 1 -C 3 haloalkyl, * and ** are each independently stereocenters, and R′, R′′, R′′′, R′′′′, and R′′′′′ are, in the order going around the aromatic ring from R′ to R′′′′′, selected from
  • R 1 is C 1 -C 3 alkyl, C 1 -C 3 deuteroalkyl, or C 1 -C 3 haloalkyl, n is an integer from 1 to 20, and * is a stereocenter; 3) Forming an addition salt of Formula (XIVb):
  • R 1 is C 1 -C 3 alkyl, C 1 -C 3 deuteroalkyl, or C 1 -C 3 haloalkyl
  • R 2 is a nitrogen protecting group
  • n is an integer from 1 to 20, and * is a stereocenter; or 4) Crystallizing a compound of Formula (I) or a salt thereof from one or more solvents, optionally chloroform and hexanes.
  • the compound of Formula (XIb) is crystallized in a mixture of tetrahydrofuran and methyl t-butyl ether.
  • the ratio of tetrahydrofuran and methyl t-butyl ether is between: 1:10 and 3:10. For example, the ratio is 1.5:10.
  • the compound of Formula (I) or a salt thereof is crystallized in a mixture of chloroform and hexanes.
  • the ratio of chloroform to hexanes is between 1:5 and 1:1.
  • the ratio is 1:3 or 1:2.
  • methods of preparing a polypeptide comprising making the polypeptide with one or more crystalline compounds of Formula (I) or their crystalline salts.
  • FIG. 1 is a chiral HPLC trace of N-Fmoc-(S)-alpha-methyl-alpha-amino-6-enoic acid.
  • FIG. 2 is a chiral HPLC trace of N-Fmoc-(S)-alpha-methyl-alpha-amino-6-enoic acid spiked with N-Fmoc-(R)-alpha-methyl-alpha-amino-6-enoic acid.
  • FIG. 3 is an HPLC trace of N-Fmoc-(S)-alpha-methyl-alpha-amino-6-enoic acid with the detector set to 215 nm.
  • FIG. 4 is an HPLC trace of N-Fmoc-(S)-alpha-methyl-alpha-amino-6-enoic acid with the detector set to 254 nm.
  • FIG. 5 is an HPLC trace of an N-Fmoc-(S)-alpha-methyl-alpha-amino-6-enoic acid standard.
  • FIG. 6 is a chiral HPLC trace of N-Fmoc-(R)-alpha-methyl-alpha-aminodec-9-enoic acid.
  • FIG. 7 is a chiral HPLC trace of N-Fmoc-(R)-alpha-methyl-alpha-aminodec-9-enoic acid spiked with N-Fmoc-(S)-alpha-methyl-alpha-aminodec-9-enoic acid.
  • FIG. 8 is an HPLC trace of N-Fmoc-(R)-alpha-methyl-alpha-aminodec-9-enoic acid with the detector set to 215 nm
  • FIG. 9 is an HPLC trace of N-Fmoc-(R)-alpha-methyl-alpha-aminodec-9-enoic acid with the detector set to 254 nm
  • ranges When ranges are present, the ranges include the range endpoints. Additionally, every subrange and value within the range is present as if explicitly written out.
  • halo or the term “halogen” each refer to fluorine, chlorine, bromine or iodine, or a radical thereof.
  • alkyl refers to a hydrocarbon chain that is a straight chain or branched chain, containing the indicated number of carbon atoms.
  • C 1 -C 3 alkyl group indicates that the group has from 1 to 3 (inclusive) carbon atoms in it.
  • Deuteroalkyl refers to a deuterated alkyl chain, where the alkyl chain hydrogen atoms are replaced at least the 90% level with deuterium atoms.
  • haloalkyl refers to a halogenated alkyl chain where the alkyl chain hydrogen atoms are replaced with halogen atoms.
  • the halogen atoms are all the same (e.g., all F or all Cl).
  • peptidomimetic macrocycle or “crosslinked polypeptide” refers to a compound comprising a plurality of amino acid residues joined by a plurality of peptide bonds and at least one macrocycle-forming linker which forms a macrocycle between a first naturally-occurring or non-naturally-occurring amino acid residue (or analog) and a second naturally-occurring or non-naturally-occurring amino acid residue (or analog) within the same molecule.
  • Peptidomimetic macrocycles include embodiments where the macrocycle-forming linker connects an ⁇ -carbon of the first amino acid residue (or analog) to the ⁇ -carbon of the second amino acid residue (or analog) in the peptide.
  • Peptidomimetic macrocycles include one or more non-peptide bonds between one or more amino acid residues and/or amino acid analog residues, and optionally include one or more non-naturally-occurring amino acid residues or amino acid analog residues in addition to any which form the macrocycle.
  • a “corresponding uncrosslinked polypeptide” when referred to in the context of a peptidomimetic macrocycle is understood to relate to a polypeptide of the same length as the macrocycle and comprising the equivalent natural amino acids of the wild-type sequence corresponding to the macrocycle.
  • amino acid refers to a molecule containing both an amino group and a carboxyl group. Suitable amino acids include, for example, both the D- and L-isomers of the naturally-occurring amino acids, as well as non-naturally occurring amino acids prepared by organic synthesis or other metabolic routes.
  • amino acid contemplates, for example, ⁇ -amino acids, natural amino acids, non-natural amino acids, and amino acid analogs.
  • ⁇ -amino acid refers to a molecule containing both an amino group and a carboxyl group bound to a carbon atom which is designated the ⁇ -carbon atom.
  • naturally occurring amino acid refers to any one of the twenty amino acids commonly found in peptides synthesized in nature, and known by the one letter abbreviations A, R, N, C, D, Q, E, G, H, I, L, K, M, F, P, S, T, W, Y and V.
  • amino acid side chain refers to a moiety attached to the ⁇ -carbon atom (or another backbone atom) in an amino acid.
  • amino acid side chain for alanine is methyl
  • amino acid side chain for phenylalanine is phenylmethyl
  • amino acid side chain for cysteine is thiomethyl
  • amino acid side chain for aspartate is carboxymethyl
  • amino acid side chain for tyrosine is 4-hydroxyphenylmethyl, etc.
  • non-naturally occurring amino acid side chains are also included, for example, those that occur in nature (e.g., an amino acid metabolite) or those that are made synthetically (e.g., an ⁇ , ⁇ di-substituted amino acid).
  • ⁇ , ⁇ di-substituted amino acid refers to a molecule or moiety containing both an amino group and a carboxyl group bound to a carbon atom (e.g., the ⁇ -carbon atom) that is also attached a natural and non-natural, to two natural, or to two non-natural amino acid side chains.
  • polypeptide can encompass two or more naturally or non-naturally-occurring amino acids joined by a covalent bond (e.g., an amide bond).
  • Polypeptides, as described herein can include full length proteins (e.g., fully processed proteins) as well as shorter amino acid sequences (e.g., fragments of naturally-occurring proteins or synthetic polypeptide fragments).
  • the term “macrocyclization reagent” or “macrocycle-forming reagent” can refer to any reagent which can be used to prepare a peptidomimetic macrocycle by mediating the reaction between two reactive olefinic groups thereon.
  • the reactive groups that, once reacted, close the linker can be for example terminal olefins (alkenes), deuterated or non-deuterated.
  • Macrocyclization reagents or macrocycle-forming reagents can be metathesis catalysts including, but not limited to, stabilized, late transition metal carbene complex catalysts such as Group VIII transition metal carbene catalysts.
  • such catalysts can contain Ru and Os metal centers having a +2 oxidation state, an electron count of 16 and pentacoordinated.
  • the catalysts can have W or Mo centers.
  • Various catalysts are disclosed in Grubbs et al., “Ring Closing Metathesis and Related Processes in Organic Synthesis” Acc. Chem. Res. 1995, 28, 446-452; U.S. Pat. No. 5,811,515; U.S. Pat. No. 7,932,397; U.S. Pat. Application Pub. No.
  • treatment is defined as the application or administration of a therapeutic agent to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient, who has a disease, a symptom of disease or a predisposition toward a disease, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disease, the symptoms of disease or the predisposition toward disease.
  • R 1 is C 1 -C 3 alkyl, C 1 -C 3 deuteroalkyl, or C 1 -C 3 haloalkyl; * is a stereocenter; n is an integer from 1 to 20; R 2 is —H or a nitrogen protecting group; and R 3 is —H or a protecting or activating group.
  • R 1 can be C 1 -C 3 alkyl.
  • R 1 can be, for example, methyl, ethyl, n-propyl, or isopropyl.
  • R 1 can be C 1 -C 3 deuteroalkyl.
  • R 1 can be, for example, —CD 3 , —CD 2 CD 3 , —CD 2 CD 2 CD 3 , or —CD(CD 3 ) 2 .
  • R 1 can be C 1 -C 3 haloalkyl.
  • the halogen can be, for example, —F, —Cl, —Br, or —I.
  • R 1 can be, for example, —CX 3 , —CX 2 CX 3 , —CX 2 CX 2 CX 3 , or —CX(CX 3 ) 2 , wherein X is a halogen.
  • R 2 can be, for example, —H, or a nitrogen protecting group selected from the group consisting of: 9-Fluorenylmethoxycarbonyl (Fmoc), Trityl (Trt), 4-Methoxytrityl (Mmt), 2-(3,5-Dimethoxyphenyl)propan-2-yloxycarbonyl (Ddz), 2-(p-Biphenylyl)-2-propyloxycarbonyl (Bpoc), 2-(4-Nitrophenylsulfonyl)ethoxycarbonyl (NSC), (1,1-Dioxobenzo[b]thiophene-2-yl)methyloxycarbonyl (Bsmoc), (1,1-Dioxonaphtho[1,2-b]thiophene-2-yl)methyloxycarbonyl ( ⁇ -Nsmoc), 1-(4,4-dimethyl-2,6-dio
  • Nitrogen protecting groups can be found, for example, in Isidro-Llobet, A., et al., “Amino Acid-Protecting Groups,” Chem. Rev. 2455-2504 (2009).
  • R 2 can be, for example, a nitrogen protecting group selected from the group consisting of 9-Fluorenylmethoxycarbonyl (Fmoc), Trityl (Trt), 4-Methoxytrityl (Mmt), 2-(3,5-dimethoxyphenyl)propan-2-yloxycarbonyl (Ddz), 2-(p-biphenylyl)-2-propyloxycarbonyl (Bpoc), 2-(4-Nitrophenylsulfonyl)ethoxycarbonyl (NSC), 1,1-Dioxobenzo[b]thiophene-2-yl)methyloxycarbonyl (Bsmoc), 1-(4,4-dimethyl-2,6-dioxocyclohex-1-ylidene)-3-methylbutyl (ivDde), Tert-butyloxycarbonyl (Boc), Benzyloxycarbonyl (Fmoc), Trityl (Trt), 4-Met
  • R 2 can be a nitrogen protecting group selected from the group consisting of 9-Fluorenylmethoxycarbonyl (Fmoc), Trityl (Trt), 4-Methoxytrityl (Mmt), 2-(3,5-dimethoxyphenyl)propan-2-yloxycarbonyl (Ddz), 2-(p-biphenylyl)-2-propyloxycarbonyl (Bpoc), Tert-butyloxycarbonyl (Boc), Benzyloxycarbonyl (Z), Allyloxycarbonyl (Alloc), 2,2,2-Trichloroethyloxycarbonyl (Troc), o-Nitrobenzenesulfonyl (oNBS), Trityl (Trt), 4-Methyltrityl (Mtt), and o-Nitrobenzenesulfonyl (O-NBS).
  • Fmoc 9-Fluorenylmethoxycarbonyl
  • R 2 can be, for example, the nitrogen protecting group 9-Fluorenylmethoxycarbonyl (Fmoc).
  • R 3 can be, for example, —H or a protecting or activating group selected from the group consisting of: tert-Butyl (tBu), 2-Chlorotrityl (2-Cl-Trt), 2,4-Dimethoxybenzyl (DMB), Benzyl (Bn), 2-Phenylisopropyl (2-PhiPr), 5-Phenyl-3,4-ethylenedioxythenyl, 9-Fluorenylmethyl (Fm), 4-(N-[1-(4,4-dimethyl-2,6-dioxocyc lohexylidene)-3-methylbutyl]-amino)benzyl (Dmab), Methyl (Me), Ethyl (Et), Carbamoylmethyl (Cam), Allyl (Al), Phenacyl (Pac), p-Nitrobenzyl (pNB), 2-
  • R 3 can be, for example —H.
  • n can range, for example, from 1-20, from 3-11, or from 3-6. n can be, for example 3 or 6 or 11. n can be 3. n can be 6. n can be 11.
  • the stereocenter * can be (R). In the crystalline compound of Formula (I) or its crystalline salt, the stereocenter * can be (S).
  • R 1 in the crystalline compound of Formula (I) or it crystalline salt, R 1 can be C 1 -C 3 alkyl; R 2 can be 9-Fluorenylmethoxycarbonyl (Fmoc); R 3 can be selected from the group consisting of —H tert-Butyl (tBu), 2-Chlorotrityl (2-cl-Trt), 2,4-Dimethoxybenzyl (DMB), Benzyl (Bn), 2-Phenylisopropyl (2-PhiPr), 5-Phenyl-3,4-ethylenedioxythenyl, 9-Fluorenylmethyl (Fm), 4-(N-[1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl]-amino)benzyl (Dmab), Methyl (Me), Ethyl (Et), Carbamoylmethyl (Cam), Ally
  • R 1 in the crystalline compound of Formula (I) or it crystalline salt, R 1 can be C 1 -C 3 alkyl; R 2 can be 9-Fluorenylmethoxycarbonyl (Fmoc); R 3 can be selected from the group consisting of —H tert-Butyl (tBu), 2-Chlorotrityl (2-cl-Trt), 2,4-Dimethoxybenzyl (DMB), Benzyl (Bn), 2-Phenylisopropyl (2-PhiPr), 5-Phenyl-3,4-ethylenedioxythenyl, 9-Fluorenylmethyl (Fm), 4-(N-[1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl]-amino)benzyl (Dmab), Methyl (Me), Ethyl (Et), Carbamoylmethyl (Cam), Ally
  • R 1 in the crystalline compound of Formula (I) or its crystalline salt, R 1 can be methyl, R 2 can be 9-Fluorenylmethoxycarbonyl (Fmoc); R 3 can be —H, n can be 3, 6, or 11, and the stereocenter * can be (R).
  • R 1 in the crystalline compound of Formula (I) or its crystalline salt, R 1 can be methyl, R 2 can be 9-Fluorenylmethoxycarbonyl (Fmoc); R 3 can be —H, n can be 3, 6, or 11, and the stereocenter * can be (S).
  • any compound, its salt, crystalline compound, or crystalline salt of a compound can have a chemical purity.
  • Chemical purity can be defined, for example, as the degree to which a substance is undiluted or unmixed with extraneous material, and can be typically expressed as a percentage.
  • Any compound, salt thereof, crystalline compound, or crystalline salt of a compound herein can have, for example, a chemical purity ranging from about 90% to 100%.
  • the chemical purity can be, for example, about 92% to 100%, about 94% to 100%, about 96% to 100%, about 98% to 100%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100%.
  • the percentage can be, for example, based on the total weight of the compound, its salt, crystalline compound, or its salt.
  • the percentage can be, for example, arrived at using HPLC.
  • the percentage can be arrived at, for example, using NMR, for example proton NMR.
  • the chemical purity can be arrived at, for example, using elemental analysis.
  • any compound, salt thereof, crystalline compound, or crystalline salt of a compound can have an enantiomeric excess.
  • the enantiomeric excess can be, for example, from about 80% to 100%, from about 85% to 100%, from about 90% to 100%, from about 95% to 100%, from about 96% to 100%, from about 97% to 100%, from about 98% to 100%, from about 99% to 100%, about 95%, about 96%, about 97%, about 97.2%, about 98%, about 99%, or 100%.
  • the enantiomeric excess can be, for example, greater than 95%, greater than 96%, greater than 97%, greater than 98%, or greater than 99%.
  • the amount (e.g., moles) or enantiomer produced can be determined, for example, by chiral HPLC, by chiral GC, or via a chiral NMR shift reagent using NMR spectroscopy.
  • any compound, its salt, crystalline compound, or crystalline salt of a compound can have an optical purity.
  • the optical purity can be, for example, from about 80% to 100%, from about 85% to 100%, from about 90% to 100%, from about 95% to 100%, about 95%, about 96%, about 97%, about 98%, about 99%, or 100%.
  • specific rotation can be defined as the observed angle of optical rotation, a, when plane-polarized light is passed through a sample with a path length of 1 decimeter and a sample concentration of 1 gram per 1 millilitre.
  • the specific rotation can be obtained, for example, at 20° C. and at a wavelength of light of 589 nanometers (e.g., the sodium D line).
  • the specific rotation can be obtained, for example, with a polarimeter.
  • the solvent the sample is dissolved in can be any suitable solvent or solvent combination, for example, ethanol, methanol, chloroform, dichloromethane, carbon tetrachloride, water, DMSO, N,N-DMF, diethyl ether, tetrahydrofuran, hexane, pentane, acetone, or any combination thereof.
  • the compounds, salts, crystalline compounds, or crystalline salts of compounds herein can be diastereomers.
  • the compounds, crystalline compounds, or crystalline salts of compounds herein can have a diastereomeric excess of, for example, from about 80% to 100%, from about 85% to 100%, from about 90% to 100%, from about 95% to 100%, about 95%, about 96%, about 97%, about 98%, about 99%, or 100%.
  • diasteromeric excess for example, in a mixture of two diastereomers
  • diastereomeric excess de % ((D1 ⁇ D2)/(D1+D2))*100%, wherein D1 represents, for example, the mole or percent weight of a first and most abundant diastereomer, and D2 represents for example, the mole or percent weight of a second and least abundant diastereomer, where mole percent is used consistently (e.g., alone) in the calculation, or where percent weight is used consistently (e.g., alone) in the calculation.
  • any compound, salt thereof, crystalline compound, or crystalline salt thereof, herein, that is a diastereomer can be converted to an enantiomer or enantiomeric mixture having one stereocenter (e.g., * in Formula (I)) by, for example, removal of a nitrogen protecting group (e.g., removal of the nitrogen protecting group R 2 in the crystalline compound of Formula (I) or its crystalline salt that, together with the stereocenter *, creates a diastereomer), and the resulting enantiomer or enantiomeric mixture can then have its enantiomeric excess or optical purity determined as described herein.
  • a nitrogen protecting group e.g., removal of the nitrogen protecting group R 2 in the crystalline compound of Formula (I) or its crystalline salt that, together with the stereocenter *, creates a diastereomer
  • the resulting enantiomeric excess or optical purity is termed a converted enantiomeric excess or converted optical purity.
  • the converted enantiomeric excess can be, for example, from about 80% to 100%, from about 85% to 100%, from about 90% to 100%, from about 95% to 100%, from about 96% to 100%, from about 97% to 100%, from about 98% to 100%, from about 99% to 100%, about 95%, about 96%, about 97%, about 97.2%, about 98%, about 99%, or 100%.
  • the converted enantiomeric excess can be, for example, greater than 95%, greater than 96%, greater than 97%, greater than 98%, or greater than 99%.
  • the converted optical purity can be, for example, from about 80% to 100%, from about 85% to 100%, from about 90% to 100%, from about 95% to 100%, about 95%, about 96%, about 97%, about 98%, about 99%, or 100%.
  • any optionally crystalline diastereomer or its optionally crystalline salt herein, unless otherwise indicated, can have a converted enantiomeric excess or converted optical purity.
  • the crystalline compound of Formula (I) or its crystalline salt can be a compound of Formula (IIa) or its crystalline salt:
  • the crystalline compound of Formula (I) or its crystalline salt can be a compound of Formula (IIb) or its crystalline salt:
  • the crystalline compound of Formula (I) or its crystalline salt can be a compound of Formula (IIIa) or its crystalline salt:
  • the crystalline compound of Formula (I) or its crystalline salt can be a compound of Formula (IIIb) or its crystalline salt:
  • each E is independently selected from the group consisting of deuterium and halogen
  • R 4 is selected from the group consisting of C 1 -C 3 alkyl, C 1 -C 3 deuteroalkyl and C 1 -C 3 haloalkyl.
  • D deuterium
  • Scheme I depicts formation of the crystalline N-Fmoc-(R)- ⁇ -methyl- ⁇ -aminodec-9-enoic acid (i.e., the crystalline compound of Formula (IIa)).
  • Sequence I starts with Boc-D-proline (i.e., the compound of Formula (V)). It is understood that by starting with Boc-L-proline, compounds with the opposite stereochemistry of the compound of Formula (IIa) can be produced (e.g., the compound of Formula (IIb) can be produced).
  • stereochemisty of the amino acid used to form the metal complex is not dispositive of the stereochemistry in the resulting crystalline compound (e.g., of Formula (IIa)) or its crystalline salt.
  • Boc-D-proline (Compound of Formula (V)) is first reacted with 2-aminobenzophenone (compound of Formula (VI)) to form the compound of Formula (VII).
  • the compound of Formula (VII) is deprotected to form the HCl salt of the compound of Formula (VIII).
  • acids other than HCl including organic acids and inorganic acids, for example, nitric acid, phosphoric acid, sulfuric acid, boric acid, hydrofloric acid, hydrobromic acid, and perchloric acid.
  • benzyl halides are found in Belokon, Y. N., et al., “Halo-substituted (S)—N-(2-benzoylphenyl)-1-benzylpyrrolidine-2-carboxamides as new chiral auxiliaries for the asymmetric synthesis of (S)- ⁇ -amino acids,” Russian Chemical Bulletin, International Edition, 51(8): 1593-1599 (2002). Further and different benzyl halides could also be employed:
  • the reaction can be run in the presence of a base, for example, a hydroxide, for example an inorganic hydroxide, for example, potassium hydroxide.
  • a base for example, a hydroxide, for example an inorganic hydroxide, for example, potassium hydroxide.
  • hydroxides are contemplated, including sodium hydroxide, cesium hydroxide, lithium hydroxide, magnesium hydroxide, and ammonium hydroxide.
  • the metal complex of Formula (XI) can be crystallized one or more times from one or more solvents, for example a cyclic ether and a non-cyclic ether.
  • the solvent is tetrahydrofuran and methyl tert-butyl ether.
  • the ratio of the cyclic ether to the non-cyclic ether is at most 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10.
  • the ratio of the cyclic ether to the non-cyclic ether is at least 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10.
  • the metal complex of Formula (XI) is crystallized from a mixture of tetrahydrofuran and methyl tert-butyl ether in ratio of at most 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10.
  • the ratio of and tetrahydrofuran and methyl tert-butyl ether is at least 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10. In some cases the ratio of tetrahydrofuran and methyl tert-butyl ether is 1.5:10.
  • the metal complex of Formula (XI) may also be crystallized with esters, for example with ethyl acetate or isopropyl acetate.
  • the product or crystallized product of Formula (IX) can alternatively or additionally be crystallized or recrystallized from a solvent, for example an alcohol, for example isopropyl alcohol.
  • alcohols including methanol, ethanol, n-propanol, a butanol, n-butanol, iso-butanol, sec-butanol, and tert-butanol.
  • the metal complex of Formula (XI) is then alkylated with 8-bromooct-1-ene to form the alkylated metal complex of Formula (XII).
  • alkylating agents including other halo alkyl olefins, could be used in place of 8-bromooct-1-ene.
  • alkylating agents of the Formula (XV) could be used:
  • X is Cl, Br, or I
  • n is an integer from 1 to 20.
  • n can be from 3 to 11, from 3 to 6, or 3 or 6.
  • Some or all of the hydrogen atoms present in the compound of Formula (XV) can be replaced with deuterium atoms or halogen atoms.
  • the alkylation can be performed in one or more solvents, for example a polar aprotic solvent, for example N,N-dimethyl formamide (DMF).
  • the alkylation can be performed, for example, at a temperature of less than 20° C., for example, from less than 20° C. to 5° C., from less than 20° C. to 10° C., or at about 10° C.
  • the first alkylation could be performed using a C 1 -C 3 alkane with a leaving group such as a halogen (e.g., methyl bromide, ethyl bromide, n-propyl bromide), or a C 1 -C 3 deuteroalkane with a leaving group such as a halogen (e.g., CD 3 Br, CD 3 CD 2 Br, CD 3 CD 2 CD 2 Br), or a C 1 -C 3 haloalkane with a leaving group such as a more reactive halogen than the other halogens in the haloalkane (e.g., CF 3 Br, CF 3 CF 2 Br, CF 3 CF 2 CF 2 Br).
  • the second alkylation could be performed using the alkylating agent of Formula (XV).
  • Purification of Formula (XII) may be achieved by crystallization one or more times from one or more solvents including cyclic and non-cyclic ethers, esters, hexanes and heptanes.
  • crystallization may be achieved using a combination of ethyl acetate and hexanes, ethyl acetate and heptanes, isopropyl acetate and hexanes, isopropyl acetate and heptanes, methyl tertiary-butyl ether and hexanes, methyl tertiary-butyl ether and heptanes or isopropyl acetate and methyl tertiary-butyl ether.
  • the metal complex of Formula (XII) is then cleaved with an acid, for example HCl, using one or more solvents, for example an ether, for example a cyclic ether, for example tetrahydrofuran, to form the amino acid HCl salt of Formula (XIII).
  • an acid for example HCl
  • solvents for example an ether, for example a cyclic ether, for example tetrahydrofuran
  • other acids in addition to HCl are contemplated, for example organic or inorganic acids, for example, nitric acid, phosphoric acid, sulfuric acid, boric acid, hydrofloric acid, hydrobromic acid, or perchloric acid.
  • the salt of Formula (XIII) may be further purified by crystallization one or more times with one or more solvents.
  • the solvent may be any suitable solvent including tetrahydrofuran, methyl tertiary-butyl ether, ethyl acetate, isopropyl acetate, ethanol, methanol, isopropanol, acetonitrile, or a combination thereof.
  • the solvent is acetonitrile.
  • the amino acid salt of Formula (XIII) is then nitrogen protected with a nitrogen protecting group, in this case an Fmoc group, and the cyclohexylamine addition salt of the protected amino acid is formed, yielding the protected amino acid cyclohexylamine salt of Formula (XIV).
  • Formation of the salt of Formula (XIV) can be achieved in any suitable solvent including acetonitrile, methyl tertiary-butyl ether, tetrahydrofuran or a combination thereof.
  • the solvent is methyl tertiary-butyl ether.
  • amines for example other cyclic amines, for example cyclopropylamine, cyclobutyl amine, cyclopentylamine, cycloheptylamine, and cyclooctylamine, are contemplated.
  • nitrogen protecting groups are contemplated, for example the nitrogen protecting groups for R 2 in the crystalline compound of Formula (I) or its crystalline salt herein.
  • the protected amino acid cyclohexylamine salt of Formula (XIV) can then be crystallized from one or more ethers, for example, two ethers, for example a cyclic ether and a non cyclic ether, for example tetrahydrofuran and methyl tert-butyl ether.
  • the crystallized amino acid cyclohexylamine salt of Formula (XIV) is then treated with sulfuric acid, and subsequently crystallized to form the crystalline compound of Formula (IIa).
  • acids other than sulfuric acid are contemplated, for example organic or inorganic acids, for example, nitric acid, phosphoric acid, sulfuric acid, boric acid, hydrofloric acid, hydrobromic acid, or perchloric acid.
  • the crystallization can be performed using one or more solvents, for example two solvents, for example an alkane and haloalkane, for example hexanes and chloroform.
  • the ratio of the alkane to the haloalkane is at least 6:1, 5:1, 4:1, 3:1, 2:1, or 1:10. In some cases the ratio of the alkane to the haloalkane is at most 6:1, 5:1, 4:1, 3:1, 2:1, or 1:10.
  • the crystalline compound of Formula (IIa) may be obtained by crystallization from a mixture of hexanes and chloroform in the ratio of at least 6:1, 5:1, 4:1, 3:1, 2:1, or 1:1.
  • the crystalised IIa may also obtained by crystallization from a mixture of hexanes and chloroform in the ratio of at most 6:1, 5:1, 4:1, 3:1, 2:1, or 1:1. In some cases the ratio of hexanes and chloroform is 3:1.
  • the crystallization can be performed at a temperature ranging from, for example, about ⁇ 5° C. to about ⁇ 20° C., about ⁇ 10° C. to about ⁇ 20° C., or about ⁇ 15° C. to ⁇ 20° C.
  • the crystalline compound of Formula (IIa) could be further activated or protected at its carboxylic acid function with, for example, a protecting or activating group R 3 of the crystalline compound of Formula (I) or its crystalline salt.
  • the compounds, their salts, crystalline compounds, and their crystalline salts, herein can be produced using exemplary Scheme II (with modifications that would be readily apparent to a skilled artisan).
  • Scheme II depicts formation of the crystalline N-Fmoc-(S)- ⁇ -methyl- ⁇ -aminohept-6-enoic acid (i.e., the crystalline compound of Formula (IIIa)).
  • Sequence II starts with Boc-L-Proline (i.e., the compound of Formula (Va)). It is understood that by starting with Boc-D-proline, compounds with the opposite stereochemistry of the compound of Formula (IIIa) can be produced (e.g., the compound of Formula (III) can be produced). It is also understood that the stereochemisty of the amino acid used to form the metal complex, and whose alpha carbon atom is subsequently alkylated by the haloolefin (e.g., alanine in Formula (XIa)) is not dispositive of the stereochemistry in the resulting crystalline compound (e.g., of Formula (III)) or its crystalline salt.
  • Boc-L-Proline i.e., the compound of Formula (Va)
  • Boc-D-proline compounds with the opposite stereochemistry of the compound of Formula (IIIa) can be produced (e.g., the compound of Formula (III) can be produced). It is also understood that the stereochemisty of the amino acid used to form the metal
  • Boc-L-proline (Compound of Formula (Va)) is first reacted with 2-aminobenzophenone (compound of Formula (VI)) to form the compound of Formula (VIIa).
  • the compound of Formula (VIIa) is deprotected to form the HCl salt of the compound of Formula (VIIIa).
  • acids other than HCl including organic acids and inorganic acids, for example, nitric acid, phosphoric acid, sulfuric acid, boric acid, hydrofloric acid, hydrobromic acid, and perchloric acid.
  • benzyl halides are found in Belokon, Y. N., et al., “Halo-substituted (S)—N-(2 benzoylphenyl)-1-benzylpyrrolidine-2-carboxamides as new chiral auxiliaries for the asymmetric synthesis of (S)- ⁇ -amino acids,” Russian Chemical Bulletin, International Edition, 51(8): 1593-1599 (2002). Further and different benzyl halides could also be employed:
  • the compound of Formula (IXa) is reacted with L-alanine and Ni(NO 3 ) 2 to form the metal complex of Formula (XIa).
  • L-alanine L-alanine
  • Ni(NO 3 ) 2 The skilled artisan would understand that other amino acids other than alanine could be employed in Scheme II.
  • glycine 2-aminobutanoic acid, 2-aminopentanoic acid, and valine could be employed, for example in their D or L forms.
  • the Ni(NO 3 ) 2 can be a hydrate, for example, a hexahydrate.
  • the reaction can be run in an alcoholic solvent, for example, methanol.
  • the reaction can be run at an elevated temperature, for example, from about 40° C. to about 60° C.
  • the reaction can be run in the presence of a base, for example, a hydroxide, for example an inorganic hydroxide, for example, potassium hydroxide.
  • a base for example, a hydroxide, for example an inorganic hydroxide, for example, potassium hydroxide.
  • hydroxides are contemplated, including sodium hydroxide, lithium hydroxide, cesium hydroxide, and magnesium hydroxide.
  • the metal complex of Formula (XIa) can be crystallized one or more times from one or more solvents, for example a cyclic ether and a non-cyclic ether, for example tetrahydrofuran and methyl tert-butyl ether.
  • solvents for example a cyclic ether and a non-cyclic ether, for example tetrahydrofuran and methyl tert-butyl ether.
  • the ratio of the cyclic ether to the non-cyclic ether is at most 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10.
  • the ratio of the cyclic ether to the non-cyclic ether is at least 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10.
  • the metal complex of Formula (XIa) is crystallized from a mixture of tetrahydrofuran and methyl tert-butyl ether in ratio of at most 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10.
  • the ratio of and tetrahydrofuran and methyl tert-butyl ether is at least 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10. In some cases the ratio of tetrahydrofuran and methyl tert-butyl ether is 1.5:10.
  • the product or crystallized product of Formula (IXa) can be crystallized or recrystallized from a solvent, for example an alcohol, for example isopropyl alcohol.
  • Alcohols are contemplated, including methanol, ethanol, n-propanol, a butanol, n-butanol, iso-butanol, sec-butanol, and t-butanol.
  • Other solvents suitable for crystallization or recrystallization of Formula (XIa) include esters, for example ethyl acetate or isopropyl acetate.
  • the metal complex of Formula (XIa) is then alkylated with 5-bromopent-1-ene to form the alkylated metal complex of Formula (XIIa).
  • alkylating agents including other halo alkyl olefins, could be used in place of 5-bromopent-1-ene.
  • alkylating agents of the Formula (XV) could be used:
  • X is Cl, Br, or I
  • n is an integer from Ito 20.
  • n can be from 3 to 11, from 3 to 6, or 3 or 6.
  • Some or all of the hydrogen atoms present in the compound of Formula (XV) can be replaced with deuterium atoms or halogen atoms.
  • the alkylation can be performed in one or more solvents, for example a polar aprotic solvent, for example N,N-dimethyl formamide (DMF).
  • the alkylation can be performed, for example, at a temperature of less than 20° C., for example, from less than 20° C. to 5° C., from less than 20° C. to 10° C., or at about 10° C.
  • the first alkylation could be performed using a C 1 -C 3 alkane with a leaving group such as a halogen (e.g., methyl bromide, ethyl bromide, n-propyl bromide), or a C 1 -C 3 deuteroalkane with a leaving group such as a halogen (e.g., CD 3 Br, CD 3 CD 2 Br, CD 3 CD 2 CD 2 Br), or a C 1 -C 3 haloalkane with a leaving group such as a more reactive halogen than the other halogens in the haloalkane (e.g., CF 3 Br, CF 3 CF 2 Br, CF 3 CF 2 CF 2 Br).
  • the second alkylation could be performed using the alkylating agent of Formula (XV).
  • Purification of Formula (XIIa) may be achieved by crystallization one or more times from one or more solvents including cyclic and non-cyclic ethers, esters, hexanes and heptanes.
  • crystallization may be achieved by using a combination of ethyl acetate and hexanes, ethyl acetate and heptanes, isopropyl acetate and hexanes, isopropyl acetate and heptanes, methyl tertiary-butyl ether and hexanes, methyl tertiary-butyl ether and heptanes or isopropyl acetate and methyl tertiary-butyl ether.
  • the metal complex of Formula (XIIa) is then cleaved with an acid, for example HCl, using one or more solvents, for example an ether, for example a cyclic ether, for example tetrahydrofuran, to form the amino acid HCl salt of Formula (XIIIa).
  • an acid for example HCl
  • solvents for example an ether, for example a cyclic ether, for example tetrahydrofuran
  • the salt of Formula (XIIIa) may be further purified by crystallization one or more times with one or more solvents.
  • the solvent may be any suitable solvent including tetrahydrofuran, methyl tertiary-butyl ether, ethyl acetate, isopropyl acetate, ethanol, methanol, isopropanol, acetonitrile, or a combination thereof.
  • the solvent is acetonitrile.
  • the amino acid salt of Formula (XIIIa) is then nitrogen protected with a nitrogen protecting group, in this case an Fmoc group, yielding the protected amino acid of Formula (XIVa).
  • a nitrogen protecting group in this case an Fmoc group
  • the compound of Formula (XIVa) is taken on to the crystallization step as is.
  • the compound of Formula (XIVa) is converted to a salt prior to crystallization. Formation of the salt of Formula (XIVa) may be achieved in any suitable solvent including acetonitrile, methyl tertiary-butyl ether, tetrahydrofuran or a combination thereof.
  • nitrogen protecting groups are contemplated, for example the nitrogen protecting groups for R 2 in the crystalline compound of Formula (I) or its crystalline salt herein.
  • a protected amino acid cyclohexylamine salt of Formula (XIVa) can then be crystallized from one or more ethers, for example, two ethers, for example a cyclic ether and a non cyclic ether, for example tetrahydrofuran and methyl tert-butyl ether.
  • the protected amino acid cyclohexylamine salt of Formula (XIVa) can then be crystallized to form the crystalline compound of Formula (IIIa).
  • the crystallization can be performed using one or more solvents, for example two solvents, for example an alkane and haloalkane, for example hexanes and chloroform.
  • the ratio of the alkane to the haloalkane is at least 6:1, 5:1, 4:1, 3:1, 2:1, or 1:10.
  • the ratio of the alkane to the haloalkane is at most 6:1, 5:1, 4:1, 3:1, 2:1, or 1:10.
  • the crystalline compound of Formula (IIIa) may be obtained by crystallization from a mixture of hexanes and chloroform in the ratio of at least 6:1, 5:1, 4:1, 3:1, 2:1, or 1:1.
  • the crystalised IIIa may also obtained by crystallization from a mixture of hexanes and chloroform in the ratio of at most 6:1, 5:1, 4:1, 3:1, 2:1, or 1:1. In some cases the ratio of hexanes and chloroform is 2:1.
  • the crystallization can be performed at a temperature ranging from, for example, about ⁇ 5° C. to about ⁇ 20° C., about ⁇ 10° C. to about ⁇ 20° C., or about ⁇ 15° C. to ⁇ 20° C.
  • any compound or its salt may be crystalline.
  • any compound or its salt may be crystalline at a temperature, for example, of about 0° C. or less, about ⁇ 5° C. or less, about ⁇ 10° C. or less, about ⁇ 15° C. or less, about ⁇ 20° C.
  • the crystalline compound of Formula (IIIa) could be further activated or protected at its carboxylic acid function with, for example, a protecting or activating group R 3 of the crystalline compound of Formula (I) or its crystalline salt.
  • the crystalline compounds and their crystalline salts of Formula (I), including the crystalline compounds and their crystalline salts of Formulae (IIa), (IIb), (IIIa) and (IIIb), as well as the optionally crystalline compounds and their optionally crystalline salts of Formula (IV), can be used to synthesize peptides, polypeptides, and crosslinked polypeptides that are useful for treating and preventing diseases.
  • the crosslinked polypeptides can contain secondary structures such as a helix, for example, an alpha helix.
  • the crosslinker can stablize the secondary structures relative to an otherwise identical but uncrosslinked polypeptide.
  • the crosslinker can be formed by, for example, joining the terminal alkene side chains of, for example, two crystalline alkene ⁇ , ⁇ -disubstituted amino acids or their crystalline salts herein that are incorporated into a polypeptide through, for example, a metal catalyzed olefin metathesis reaction (e.g., forming a stapled peptide). This process is depicted in Scheme III, below:
  • stapled polypeptides are found, inter alia, for example, in International Application No. PCT/US2004/038403.
  • the crystalline compounds and their crystalline salts of Formula (I), including the crystalline compounds and their crystalline salts of Formulae (IIa), (IIb), (IIIa) and (IIIb), as well as the optionally crystalline compounds and their optionally crystalline salts of Formula (IV), can be used to synthesize peptides, polypeptides, and stitched polypeptides that are useful for treating and preventing diseases.
  • two of the crystalline compounds and their crystalline salts of Formula (I), can be incorporated into a polypeptide backbone along with an ⁇ , ⁇ -disubstituted amino acid having terminal olefins on each of its side chains, for example the compound of Formula (XVI):
  • stitched polypeptides are found, for example, in International Application Publication No. WO2008/121767.
  • peptide synthesis encompasses coupling of two or more amino acids with the aid of a coupling reagent.
  • Peptide synthesis may be performed in “liquid” or “solution” phase where the coupling of the amino acids is performed in a solvent system.
  • Peptide synthesis may also, or alternatively, be performed on “solid phase” where an amino acid is attached to a polymeric or solid support by a covalent bond at the N- or C-terminus of an amino acid.
  • Peptides can be made, for example, by chemical synthesis methods, such as those described in Fields et al., Chapter 3 in Synthetic Peptides: A User's Guide , ed. Grant, W. H.
  • peptides can be synthesized using automated Merrifield techniques of solid phase synthesis with the amino groups of the amino acids employed in the synthesis protected, for example by t-Boc or Fmoc protecting groups.
  • An automated peptide synthesizer e.g., Applied Biosystems (Foster City, Calif.), Model 430A, 431, or 433 can be employed in making peptides.
  • peptidomimetic precursors and peptidomimetic macrocycles and their salts described herein can be produced using solid phase peptide synthesis (SPPS), where for example, a C-terminal amino acid is attached to a cross-linked polystyrene resin via an acid or base labile bond with a linker.
  • SPPS solid phase peptide synthesis
  • the resin can be, for example, insoluble in the solvents used for synthesis, making it relatively simple and fast to wash away excess reagents and by-products.
  • the N-terminus of each amino acid added to the growing peptide chain can be protected, for example, with an Fmoc group, which is stable in acid, but removable by base. Side chain functional groups can be protected, as necessary or desirable, for example, with base stable, acid labile groups.
  • the peptidomimetic precursors can be made, for example, in a high-throughput, combinatorial fashion using, for example, a high-throughput polychannel combinatorial synthesizer (e.g., Thuramed TETRAS multichannel peptide synthesizer from CreoSalus, Louisville, Ky. or Model Apex 396 multichannel peptide synthesizer from AAPPTEC, Inc., Louisville, Ky.).
  • a high-throughput polychannel combinatorial synthesizer e.g., Thuramed TETRAS multichannel peptide synthesizer from CreoSalus, Louisville, Ky. or Model Apex 396 multichannel peptide synthesizer from AAPPTEC, Inc., Louisville, Ky.
  • solution peptide synthesis can be performed in a manner wherein reagents are fully or partially dissolved in, for example, an appropriate solvent, for example, a polar aprotic solvent.
  • an appropriate solvent for example, a polar aprotic solvent.
  • a solid crystalline N-terminally protected olefinic amino acid with a removable protecting group e.g., t-Butyloxycarbonyl, Benzyloxycarbonyl, Fluorenylmethoxycarbonyl
  • a C-protected amino acid with a selectively removable ester e.g., methyl, benzyl, t-butyl
  • the amino acids can be fully or partially dissolved in a solvent and an activating agent is added to accomplish peptide bond formation between the amino acids.
  • Solution peptide synthesis can also utilize first formation of active esters of N-protected olefinic amino acids (e.g., N-hydroxysuccinamide, p-nitrophenyl, 2,4,6-trichlorophenyl, pentafluorophenyl) and then subsequent reaction of the activated amino acid with an unprotected or C-protected amino acid.
  • the active esters of olefinic amino acids can be prepared, for example, by reacting a solid N-protected olefinic amino acid with an appropriate alcohol with help of the condensing agent (e.g., dicyclohexylcarbodiimide).
  • the condensing agent e.g., dicyclohexylcarbodiimide
  • Formation of C-terminally protected olefinic amino acids can easily be facilitated by reacting dry solid olefinic amino acid(s) with an appropriate alcohol (e.g., methyl, ethyl, benzyl) under, for example, anhydrous conditions. Formation of a peptide where olefinic amino acid is located in the C-terminal position can accomplished, for example, in the similar way. Solution methods of peptide preparation can be easily adapted to process scale.
  • an appropriate alcohol e.g., methyl, ethyl, benzyl
  • the starting materials and intermediates of the reactions of any embodiment herein, herein and as-above disclosed, unless otherwise indicated, may be isolated and purified if desired using conventional techniques, including, but not limited to filtration, distillation, crystallization, chromatogram, flash chromotography, HPLC, MPLC, Chromatotron®, ion exchange chromatography, crystallization with Mosher acids or Mosher esters, and the like.
  • Such materials may be characterized using conventional means, including physical constructs and spectral data, for example proton NMR, carbon NMR, IR spectroscopy, polarimetry, atomic absorption, elemental analysis, UV spectroscopy, FTIR spectroscopy, and the like.
  • chromatography can be excluded in making any of the compounds or their salts.
  • the reactions described herein can take place at, for example, from about 0.001 to about 100 atmospheres (atm), for example, about 0.001 atm, about 0.01 atm, about 0.1 atm, about 1 atm, about 2 atm, about 3 atm, about 4 atm, about 5 atm, about 10 atm, about 20 atm, about 50 atm, or about 100 atm.
  • Reactions in any embodiment herein, unless otherwise indicated, can be run, unless otherwise specified, for example, open to the atmosphere, or under an inert gas atmosphere such as, for example, nitrogen or argon.
  • Reactions in any embodiment herein, unless otherwise indicated, can be run, unless otherwise specified, for example, at temperatures from about ⁇ 78° C. to about 150° C., for example from about ⁇ 78° C., about ⁇ 50° C., about ⁇ 20° C., about 0° C., about 10° C., about 20° C., about 23° C., about 25° C., about 27° C., about 30° C., about 40° C., about 50° C., about 100° C., about 125° C., about 150° C., at about ambient temperature, or at about room temperature.
  • Reactions herein, unless otherwise indicated, can have a yield, unless otherwise explicitly stated, based on the theoretical yield, for example, ranging from about 1% to about 99%.
  • the yield can be, for example, about 99%, about 98%, about 97%, about 96%, about 95%, about 90%, about 85%, about 80%, about 75%, about 70%, about 65%, about 60%, about 55%, about 50%, about 45%, about 40%, about 35%, about 30%, about 25%, about 20%, about 15%, about 10%, or about 5%.
  • Reactions herein, unless otherwise indicated, can be run, unless otherwise specified, for example, for a time ranging from about 0.1 to about 96 hours, e.g., for about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 13 hours, about 14 hours, about 15 hours, about 16 hours, about 17 hours, about 18 hours, about 19 hours, about 20 hours, about 21 hours, about 22 hours, about 23 hours, about 24 hours, about 48 hours, about 72 hours, or about 96 hours,
  • Crosslinked peptidomimetic macrocycles (stitched or stapled peptides), made with for example at least one of the crystalline compounds and their crystalline salts of Formula (I), including the crystalline compounds and their salts of Formulae (IIa), (IIb), (IIIa) and (IIIb), as well as the optionally crystalline compounds and their optionally crystalline salts of Formula (IV), can be used to treat or prevent diseases.
  • the crosslinked peptidomimetic macrocycles (stitched or stapled peptides) can be used to treat or prevent cancers.
  • cancers include, for example, fibrosarcoma, myosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, gastric cancer, esophageal cancer, rectal cancer, pancreatic cancer, ovarian cancer, prostate cancer, uterine cancer, cancer of the head and neck, skin cancer, brain cancer, squamous cell carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinom
  • Tetrahydrofuran and 9.6 kg (1.0 equivs.) of Boc-D-proline (V) were added to a reactor and cooled to ⁇ 5° C.
  • 5.3 kg (1.15 equivs.) of N-methylmorpholine were charged followed by a slow addition of 6.1 kg (1.0 equivs.) of isobutyl chloroformate in tetrahydrofuran while maintaining the internal temperature at ⁇ 5° C.
  • the mixture was allowed to agitate at 20-25° C. for 45-60 minutes and then was analyzed by TLC for completion.
  • THF was added to the crude product (15 mL per 10 g of starting material (D-BPB)) and the resulting mixture was heated to 50° C. The mixture was maintained at 50° C. for 1 h, then methyl tertiary-butyl ether was added (50 mL per 10 g of starting material (D-BPB)). The mixture was maintained at 50° C. for additional 1 h after which it was cooled to 35° C. The mixture was filtered and the resulting solid was washed with methyl tertiary-butyl ether (20 mL per 10 g of starting material (D-BPB)) to obtain the crystalline product XI.
  • the R8-Ni-BPB (XII)/DCM solution was charged to a 50-L chem-glass reactor and stripped to an oil.Tetrahydrofuran was then added and the mixture was agitated at 20-25° C. until a solution formed. 7.8 kg (5.0 equvs.) of 32% hydrochloric acid was charged slowly while maintaining an internal temperature of ⁇ 30° C. The mixture was then allowed to agitate for 6-8 hours at ambient temperature. The mixture was then concentrated to remove tetrahydrofuran to yield a slurry. Additional water was added and the slurry was agitated at ambient temperature for 1-2 hours.
  • the solid BPB salts were isolated by filtration and rinsed with additional water followed by methyl tert-butyl ether.
  • the product filtrates were then re-charged to the reactor yielding a tri-phased solution.
  • the lower-most layer was split from the upper two layers.
  • the combined two organic layers were then washed 3 ⁇ with water and concentrated to an oil.
  • Acetonitrile was then added and the mixture was warmed to 70° C. for 30 minutes.
  • the mixture was then cooled to 25-30° C. and the solid product was isolated.
  • the solid filter-cake was washed with acetonitrile and methyl tert-butyl ether, then analyzed for purity.
  • Acetonitrile (23 mL per 10 g of starting material (oil of (R)-Ala-Ni-BPB (XI)) was added to the crude product and the resulting mixture was heated to 70° C. for 30 min after which it was cooled to 20° C. The mixture was filtered and the resulting solid was washed with acetonitrile (5 mL) and methyl tertiary-butyl ether (8.5 mL) to obtain the crystalline product XIII.
  • Acetonitrile (30 mL per 10 g of starting material (oil of (R)-Ala-Ni-BPB (XI)) was added to the crude product and the resulting mixture was heated at 60° C. for 30 min followed by cooling to 30° C. The mixture was then filtered and washed with 5 mL acetonitrile to obtain the crystalline product XIII.
  • Acetonitrile (23 mL per 10 g of starting material (oil of (R)-Ala-Ni-BPB (XI)) was added to the crude product and the resulting mixture was heated at 40° C. for 30 min followed by cooling to room temperature. The mixture was then filtered and washed twice with 5 mL acetonitrile to obtain the crystalline product XIII.
  • the slurry was agitated at ambient temperature (20-25° C.) for 3 hours and the solid product salt (XIV) was isolated by filtration.
  • the solids were rinsed twice with additional methyl tert-butyl ether and the solid wetcake was recharged to a clean reactor.
  • the wetcake was recrystallized from tetrahydrofuran and methyl tert-butyl ether to improve the purity.
  • the solid salt was suspended in methyl tert-butyl ether and water and the pH adjusted to 2.0-2.5 with 25% sulfuric acid.
  • the organic product layer was washed with water until all of the cyclohexylamine was removed.
  • the organic product layer was concentrated and co-stripped with hexanes to a loose oil.
  • the product (IIa) was then crystallized out of chloroform and hexanes and dried at ⁇ 0° C. under a 1.0 cfm nitrogen sweep. Yield: 1.12 kg, 41.5%
  • Methyl tertiary-butyl ether (800 mL per 36 g of starting material XIII) was added to the crude product and the pH of the resulting mixture was adjusted to 8-9 using CHA at 20° C. The mixture was mixed at 20° C. and after 1 h crystals started forming. Additional methyl tertiary-butyl ether was added (200 mL) and the resulting slurry was mixed for 18 h. The mixture was filtered and the resulting solid was washed with twice methyl tertiary-butyl ether (200 mL and 8.5 mL) to obtain the crystalline product XIII.
  • the product was analyzed for chiral purity, and if the results were less than 95% Fmoc-R8 vs. Fmoc-S8 then crystallization was performed to upgrade the chiral purity by dissolving dry FmocR/S (50 g) in THF (50 mL). Once FmocR/S was dissolved, methyl tertiary-butyl ether was added (900 mL) and the mixture was mixed at 20° C. for 18 h. The mixture was then filtered and washed twice with methyl tertiary-butyl ether (100 mL each). The chiral purity of the resulting crystalline product XIV was about 97.8%
  • Methyl tertiary-butyl ether (1500 mL per 47 g of starting material XIII) was added to the crude product and the pH of the resulting mixture was adjusted to 8-9 using CHA at 20° C. The mixture was mixed at this temperature for 3 h after which it was filtered and the resulting solid was washed with methyl tertiary-butyl ether (250 mL).
  • Methyl tertiary-butyl ether 400 mL per 20 g of starting material XIII was added to the crude product and the pH of the resulting mixture was adjusted to 8-9 using CHA at 20° C. Additional 200 mL methyl tertiary-butyl ether was added and the mixture was mixed at this temperature for 2 h after which it was filtered and the resulting solid was washed with methyl tertiary-butyl ether (10 mL).
  • Methyl tertiary-butyl ether (50 mL per 4 g of starting material XIII) was added to the crude product and the pH of the resulting mixture was adjusted to 8-9 using CHA at 20° C. The mixture was mixed at this temperature for 45 min after which it was filtered and the resulting solid was washed with methyl tertiary-butyl ether (10 mL).
  • Chloroform (2200 m L) was added to the crude product (1100 g). Hexanes (6600 L) were then added slowly and the resulting mixture was cooled to less than 0° C. The mixture was further mixed at temperature below 0° C. for 1 h after which it was filtered at less than 0° C. and the resulting solid was dried under vacuum at temperature below 0° C.
  • Tetrahydrofuran and 7.5 kg (1.0 equivs.) of Boc-L-proline (Va) were added to a reactor and the resulting solution was cooled to ⁇ 5° C.
  • 4.2 kg (1.05 equivs.) of N-methylmorpholine were charged, followed by slow addition of 5.3 kg (1.0 equivs.) of isobutyl chloroformate in tetrahydrofuran while maintaining an internal temperature of ⁇ 5° C.
  • the mixture was allowed to agitate at 20-25° C. for 45-60 minutes and then was analyzed by TLC for completion.
  • Methyl tertiary-butyl ether (550 mL per 50 g of starting material L-BPB) was added to the crude product (S)-Ala-Ni-BPB and the slurry was then heated to 50° C. before cooling it to 20° C. The mixture was mixed at 20° C. for 16 h. The mixture was filtered and the resulting solid was washed with methyl tertiary-butyl ether (100 mL) to obtain the crystalline product XIa.
  • Methyl tertiary-butyl ether (600 mL per 50 g of starting material L-BPB) was added to the crude product (S)-Ala-Ni-BPB and the slurry was then heated to 50-60° C. and maintained at this temperature for 1 h. The mixture was then filtered at 35° C. and washed with methyl tertiary-butyl ether (100 mL) to obtain the crystalline product XIa.
  • Methyl tertiary-butyl ether (500 mL per 50 g of starting material L-BPB) was added to the crude product (S)-Ala-Ni-BPB and the slurry was then heated to 45-50° C. and maintained at this temperature for 1 h. The mixture was then filtered at 35° C. and washed with methyl tertiary-butyl ether (100 mL) to obtain the crystalline product XIa.
  • Methyl tertiary-butyl ether (2000 mL per 280 g of starting material L-BPB) was added to the crude product (S)-Ala-Ni-BPB and the slurry was then heated to 45-50° C. and maintained at this temperature for 30 min. The mixture was then cooled to 20° C. and mixed at this temperature for 8 h. The resulting solid was then filtered and washed with methyl tertiary-butyl ether (100 mL).
  • Methyl tertiary-butyl ether (100 mL per 12.5 g of starting material XIa) was added to the crude product S5-Ni-BPB and the mixture was heated to 45-50° C. Heptanes (400 mL) were added 45-50° C. The resulting slurry was cooled to 20° C. and filtered to obtain the crystalline product XIIa.
  • the solid BPB salts were isolated by filtration and rinsed with additional water followed by methyl tert-butyl ether.
  • the product filtrates were then re-charged to the reactor yielding a tri-phased solution.
  • the lower-most layer was split from the upper two layers.
  • the combined two organic layers were then washed 3 ⁇ with water and concentrated to an oil.
  • Acetonitrile was added and the mixture was warmed to 70° C. for 30 minutes.
  • the mixture was then cooled to 25-30° C. and the solid product was isolated.
  • the solid filter-cake was washed with acetonitrile and methyl tert-butyl ether, then analyzed for chemical purity.
  • Acetonitrile 100 mL per 20 g of starting material S5-Ni-BPB (XIIa) was added to the crude product and the mixture was mixed at 20° C. for 1 h. The mixture was then filtered and washed with acetonitrile (40 mL) to obtain the crystalline product XIIIa.
  • Acetonitrile 500 mL per 185 g of starting material XIIa was added to the crude product S5-Ni-BPB and the slurry was dissolved at 45-50° C. The solvent was removed under vacuum at 45-50° C., 500 mL acetonitrile was added and the resulting mixture was heated to 45-50° C. The mixture was then cooled to 35° C., filtered and washed with acetonitrile (50 mL) to obtain the crystalline product XIIIa.
  • Acetonitrile (270 mL per 35 g of starting material XIIa) was added and the slurry was heated to 45-50° C.
  • the mixture was allowed to react at 20-25° C., while maintaining the pH between 8.5-9.0 with additional amounts of the 20% sodium carbonate solution until the reaction was complete.
  • the mixture was pH adjusted down to pH 2.0-2.5 with conc. hydrochloric acid. Tetrahydrofuran was distilled off and methyl tert-butyl ether is charged. The layers were separated and the organic layer was washed 3 more times with additional water. The organic layer was then concentrated under vacuum and co-stripped with methyl tert-butyl ether. The organic product layer was concentrated and co-stripped with hexanes to a loose oil. The product (IIIa) was then crystallized out of chloroform and hexanes and dried at ⁇ 0° C. under a 1.0 cfm nitrogen sweep.
  • Methyl tertiary-butyl ether (200 mL per 5 g of starting material XIIIa) was added to the crude product and the pH was adjusted to 8-9 using cyclohexylamine at 20° C. The resulting mixture was mixed at 20° C. for 1 h and then filtered and washed with methyl tertiary-butyl ether (50 mL) to obtain the crystalline cyclohexylamine salt of IIIa.

Abstract

Provided are crystalline α, α-disubstituted amino acids and their crystalline salts containing a terminal alkene on one of their side chains, as well as optionally crystalline halogenated and deuterated analogs of the α, α-disubstituted amino acids and their salts; methods of making these, and methods of using these.

Description

    CROSS-REFERENCE
  • This application claims the benefit of U.S. Provisional Application No. 61/721,457, filed Nov. 1, 2012, and U.S. Provisional Application No. 61/799,917, filed Mar. 15, 2013, each of which is incorporated herein by reference in its entirety.
  • BACKGROUND OF THE INVENTION
  • α, α-Disubstituted amino acids bearing a terminal alkene on one of their side chains and their salts (“alkene α, α-disubstituted amino acids”) are useful for making cross-linked macrocyclic peptides. For example, International Application No. PCT/US2004/038403 (“the '403 application”) discloses incorporating into a peptide two α, α-disubstituted amino acids that each contain a side-chain bearing a terminal alkene, and cross-linking the terminal alkene groups to form a cross-linked (“stapled”) macrocyclic peptide. The cross-link can, for example, stabilize a secondary structure (e.g., an α-helix) present in the stapled macrocyclic peptide.
  • International Application Publication No. WO2008/121767 (“the '767 publication”) discloses using alkene α, α-disubstituted amino acids to form stitched polypeptides (e.g., multiple and tandem crosslinked polypeptides) having secondary structures stabilized by stitching. The '403 application, the '767 publication, and other applications, publications, and patents, disclose that stapled and stitched macrocyclic peptides are useful for treating and preventing various diseases including cancer.
  • Alkene α, α-disubstituted amino acids are thus important and useful building blocks for forming stitched and stapled polypeptides and their precursors. The use of alkene α, α-disubstituted amino acids, however, has been limited by an inability to provide these important molecules in crystalline form. For example, commercially available preparations of alkene α, α-disubstituted amino acids are typically sold as pre-made solutions. The pre-made solutions limit the amount of α, α-disubstituted amino acid that can be shipped per unit volume, limit the chemical reactions that are available to be run with the alkene α, α-disubstituted amino acids, subject the alkene α, α-disubstituted amino acids to an enhanced degradation rate, and are environmentally unfriendly. Thus, there remains a compelling need for crystalline alkene α, α-disubstituted amino acids and their crystalline salts, and processes for producing and using these crystalline amino acids.
  • In addition, substituting one or more hydrogen atoms of an alkene α, α-disubstituted amino acid with deuterium or a halogen atom can change one or more of the amino acid's properties. For example dipole moment, hydrophobicity, hydrophilicity, steric bulk, or reactivity of an alkene α, α-disubstituted amino acid can be changed by substituting one or more hydrogen atoms thereon with one or more deuterium or halogen atoms. Thus, there also remains a need for optionally crystalline alkene α, α-disubstituted amino acids and their optionally crystalline salts having one or more hydrogen atoms thereon substituted with deuterium or halogen, and methods of making and using these.
  • SUMMARY OF THE INVENTION
  • The above needs, and others, are addressed herein. The inventive embodiments provided in this Summary of the Invention are meant to be illustrative only and to provide an overview of selected inventive embodiments disclosed herein. The Summary of the Invention, being illustrative and selective, does not limit the scope of any claim, does not provide the entire scope of inventive embodiments disclosed or contemplated herein, and should not be construed as limiting or constraining the scope of this disclosure or any claimed inventive embodiment.
  • Provided herein are crystalline compounds of Formula (I) and crystalline salts thereof:
  • Figure US20140128581A1-20140508-C00001
  • wherein R1 is C1-C3 alkyl, C1-C3 deuteroalkyl, or C1-C3 haloalkyl; * is a stereocenter; n is an integer from 1 to 20; R2 is —H or a nitrogen protecting group; and R3 is —H or a protecting or activating group.
  • Also provided herein are methods of preparing a polypeptide, comprising making the polypeptide with one or more crystalline compounds of Formula (I) or their crystalline salts.
  • Further provided herein are methods of making crystalline compounds of Formula (I) or their crystalline salts, comprising at least one of the following purifications:
  • 1) Crystallizing a metal complex of Formula (XIb)
  • Figure US20140128581A1-20140508-C00002
  • from one or more solvents, optionally a cyclic ether, optionally tetrahydrofuran and methyl tert-butyl ether, or optionally an alcohol, optionally isopropyl alcohol, optionally an ester, optionally isopropyl acetate, optionally ethyl acetate, wherein R1 is C1-C3 alkyl, C1-C3 deuteroalkyl, or C1-C3 haloalkyl, * and ** are each independently stereocenters, and R′, R″, R′″, R″″, and R′″″ are, in the order going around the aromatic ring from R′ to R′″″, selected from
      • H, H, Cl, Cl, H;
      • F, F, F, F, F;
      • F, F, OiPr, F, F;
      • F, F, OMe, F, F;
      • Cl, H, H, H, H; or
      • H, H, Me, Me, H;
        2) Precipitating a compound of Formula (Ia) as its HCl salt:
  • Figure US20140128581A1-20140508-C00003
  • wherein R1 is C1-C3 alkyl, C1-C3 deuteroalkyl, or C1-C3 haloalkyl, n is an integer from 1 to 20, and * is a stereocenter;
    3) Forming an addition salt of Formula (XIVb):
  • Figure US20140128581A1-20140508-C00004
  • wherein R1 is C1-C3 alkyl, C1-C3 deuteroalkyl, or C1-C3 haloalkyl, R2 is a nitrogen protecting group, n is an integer from 1 to 20, and * is a stereocenter; or
    4) Crystallizing a compound of Formula (I) or a salt thereof from one or more solvents, optionally chloroform and hexanes.
  • In some embodiments, the compound of Formula (XIb) is crystallized in a mixture of tetrahydrofuran and methyl t-butyl ether. In some embodiments, the ratio of tetrahydrofuran and methyl t-butyl ether is between: 1:10 and 3:10. For example, the ratio is 1.5:10.
  • In some embodiments, the compound of Formula (I) or a salt thereof is crystallized in a mixture of chloroform and hexanes. In some embodiments, the ratio of chloroform to hexanes is between 1:5 and 1:1. For example, the ratio is 1:3 or 1:2. Also provided herein are methods of preparing a polypeptide, comprising making the polypeptide with one or more crystalline compounds of Formula (I) or their crystalline salts.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a chiral HPLC trace of N-Fmoc-(S)-alpha-methyl-alpha-amino-6-enoic acid.
  • FIG. 2 is a chiral HPLC trace of N-Fmoc-(S)-alpha-methyl-alpha-amino-6-enoic acid spiked with N-Fmoc-(R)-alpha-methyl-alpha-amino-6-enoic acid.
  • FIG. 3 is an HPLC trace of N-Fmoc-(S)-alpha-methyl-alpha-amino-6-enoic acid with the detector set to 215 nm.
  • FIG. 4 is an HPLC trace of N-Fmoc-(S)-alpha-methyl-alpha-amino-6-enoic acid with the detector set to 254 nm.
  • FIG. 5 is an HPLC trace of an N-Fmoc-(S)-alpha-methyl-alpha-amino-6-enoic acid standard.
  • FIG. 6 is a chiral HPLC trace of N-Fmoc-(R)-alpha-methyl-alpha-aminodec-9-enoic acid.
  • FIG. 7 is a chiral HPLC trace of N-Fmoc-(R)-alpha-methyl-alpha-aminodec-9-enoic acid spiked with N-Fmoc-(S)-alpha-methyl-alpha-aminodec-9-enoic acid.
  • FIG. 8 is an HPLC trace of N-Fmoc-(R)-alpha-methyl-alpha-aminodec-9-enoic acid with the detector set to 215 nm
  • FIG. 9 is an HPLC trace of N-Fmoc-(R)-alpha-methyl-alpha-aminodec-9-enoic acid with the detector set to 254 nm
  • INCORPORATION BY REFERENCE
  • All publications, patents, and patent applications referenced herein are incorporated by reference in their entireties. In the event of a conflict between a term herein and a term incorporated by reference, the term herein controls.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The details of one or more inventive embodiments are set forth in the accompanying drawings, the claims, and in the description herein. Other features, objects, and advantages of inventive embodiments disclosed and contemplated herein will be apparent from the description and drawings, and from the claims.
  • INITIAL DEFINITIONS
  • As used herein, unless otherwise indicated, the article “a” means one or more unless explicitly otherwise provided for.
  • As used herein, unless otherwise indicated, terms such as “contain,” “containing,” “include,” “including,” and the like mean “comprising.”
  • As used herein, unless otherwise indicated, the term “or” can be conjunctive or disjunctive.
  • Herein, unless otherwise indicated, any embodiment can be combined with any other embodiment.
  • Herein, unless otherwise indicated, some inventive embodiments herein contemplate numerical ranges.
  • When ranges are present, the ranges include the range endpoints. Additionally, every subrange and value within the range is present as if explicitly written out.
  • Herein, unless otherwise indicated, the symbol “D” stands for deuterium or a radical thereof.
  • Herein, unless otherwise indicated, the term “halo” or the term “halogen” each refer to fluorine, chlorine, bromine or iodine, or a radical thereof.
  • Herein, unless otherwise indicated, the term “alkyl” refers to a hydrocarbon chain that is a straight chain or branched chain, containing the indicated number of carbon atoms. For example, C1-C3 alkyl group indicates that the group has from 1 to 3 (inclusive) carbon atoms in it.
  • “Deuteroalkyl” refers to a deuterated alkyl chain, where the alkyl chain hydrogen atoms are replaced at least the 90% level with deuterium atoms.
  • Herein, unless otherwise indicated, the term “haloalkyl” refers to a halogenated alkyl chain where the alkyl chain hydrogen atoms are replaced with halogen atoms. In some embodiments, the halogen atoms are all the same (e.g., all F or all Cl).
  • Herein, unless otherwise indicated,
    Figure US20140128581A1-20140508-P00001
    is a double (e.g., alkene) bond.
  • As used herein, unless otherwise indicated, the term “peptidomimetic macrocycle” or “crosslinked polypeptide” refers to a compound comprising a plurality of amino acid residues joined by a plurality of peptide bonds and at least one macrocycle-forming linker which forms a macrocycle between a first naturally-occurring or non-naturally-occurring amino acid residue (or analog) and a second naturally-occurring or non-naturally-occurring amino acid residue (or analog) within the same molecule. Peptidomimetic macrocycles include embodiments where the macrocycle-forming linker connects an α-carbon of the first amino acid residue (or analog) to the α-carbon of the second amino acid residue (or analog) in the peptide. Peptidomimetic macrocycles include one or more non-peptide bonds between one or more amino acid residues and/or amino acid analog residues, and optionally include one or more non-naturally-occurring amino acid residues or amino acid analog residues in addition to any which form the macrocycle.
  • As used herein, unless otherwise indicated, a “corresponding uncrosslinked polypeptide” when referred to in the context of a peptidomimetic macrocycle is understood to relate to a polypeptide of the same length as the macrocycle and comprising the equivalent natural amino acids of the wild-type sequence corresponding to the macrocycle.
  • As used herein, unless otherwise indicated, the term “amino acid” refers to a molecule containing both an amino group and a carboxyl group. Suitable amino acids include, for example, both the D- and L-isomers of the naturally-occurring amino acids, as well as non-naturally occurring amino acids prepared by organic synthesis or other metabolic routes. The term amino acid contemplates, for example, α-amino acids, natural amino acids, non-natural amino acids, and amino acid analogs.
  • As used herein, unless otherwise indicated, the term “α-amino acid” refers to a molecule containing both an amino group and a carboxyl group bound to a carbon atom which is designated the α-carbon atom.
  • As used herein, unless otherwise indicated, the term “naturally occurring amino acid” refers to any one of the twenty amino acids commonly found in peptides synthesized in nature, and known by the one letter abbreviations A, R, N, C, D, Q, E, G, H, I, L, K, M, F, P, S, T, W, Y and V.
  • Herein, unless otherwise indicated, the term “amino acid side chain” refers to a moiety attached to the α-carbon atom (or another backbone atom) in an amino acid. For example, the amino acid side chain for alanine is methyl, the amino acid side chain for phenylalanine is phenylmethyl, the amino acid side chain for cysteine is thiomethyl, the amino acid side chain for aspartate is carboxymethyl, the amino acid side chain for tyrosine is 4-hydroxyphenylmethyl, etc. Other non-naturally occurring amino acid side chains are also included, for example, those that occur in nature (e.g., an amino acid metabolite) or those that are made synthetically (e.g., an α,α di-substituted amino acid).
  • Herein, unless otherwise indicated, the term “α,αdi-substituted amino” acid refers to a molecule or moiety containing both an amino group and a carboxyl group bound to a carbon atom (e.g., the α-carbon atom) that is also attached a natural and non-natural, to two natural, or to two non-natural amino acid side chains.
  • Herein, unless otherwise indicated, the term “polypeptide” can encompass two or more naturally or non-naturally-occurring amino acids joined by a covalent bond (e.g., an amide bond). Polypeptides, as described herein can include full length proteins (e.g., fully processed proteins) as well as shorter amino acid sequences (e.g., fragments of naturally-occurring proteins or synthetic polypeptide fragments).
  • Herein, unless otherwise indicated, the term “macrocyclization reagent” or “macrocycle-forming reagent” can refer to any reagent which can be used to prepare a peptidomimetic macrocycle by mediating the reaction between two reactive olefinic groups thereon. The reactive groups that, once reacted, close the linker, can be for example terminal olefins (alkenes), deuterated or non-deuterated.
  • Macrocyclization reagents or macrocycle-forming reagents can be metathesis catalysts including, but not limited to, stabilized, late transition metal carbene complex catalysts such as Group VIII transition metal carbene catalysts. For example, such catalysts can contain Ru and Os metal centers having a +2 oxidation state, an electron count of 16 and pentacoordinated. The catalysts can have W or Mo centers. Various catalysts are disclosed in Grubbs et al., “Ring Closing Metathesis and Related Processes in Organic Synthesis” Acc. Chem. Res. 1995, 28, 446-452; U.S. Pat. No. 5,811,515; U.S. Pat. No. 7,932,397; U.S. Pat. Application Pub. No. 2011/0065915; U.S. Pat. Application Pub. No. 2011/0245477; Yu et al., “Synthesis of Macrocyclic Natural Products by Catalyst-Controlled Stereoselective Ring-Closing Metathesis,” Nature 2011, 479, 88; and Peryshkov et al., “Z-Selective Olefin Metathesis Reactions Promoted by Tungsten Oxo Alkylidene Complexes,” J. Am. Chem. Soc. 2011, 133, 20754.
  • Herein, unless otherwise indicated, the term “treatment” is defined as the application or administration of a therapeutic agent to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient, who has a disease, a symptom of disease or a predisposition toward a disease, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disease, the symptoms of disease or the predisposition toward disease.
  • Provided herein are crystalline compounds of Formula (I) or their crystalline salts:
  • Figure US20140128581A1-20140508-C00005
  • wherein R1 is C1-C3 alkyl, C1-C3 deuteroalkyl, or C1-C3 haloalkyl; * is a stereocenter; n is an integer from 1 to 20; R2 is —H or a nitrogen protecting group; and R3 is —H or a protecting or activating group.
  • R1
  • In the crystalline compound of Formula (I) or its crystalline salt, R1 can be C1-C3 alkyl. R1 can be, for example, methyl, ethyl, n-propyl, or isopropyl.
  • In the crystalline compound of Formula (I) or its crystalline salt, R1 can be C1-C3 deuteroalkyl. R1 can be, for example, —CD3, —CD2CD3, —CD2CD2CD3, or —CD(CD3)2.
  • In the crystalline compound of Formula (I) or its crystalline salt, R1 can be C1-C3 haloalkyl. The halogen can be, for example, —F, —Cl, —Br, or —I. R1 can be, for example, —CX3, —CX2CX3, —CX2CX2CX3, or —CX(CX3)2, wherein X is a halogen.
  • R2
  • In the crystalline compound of Formula (I) or its crystalline salt, R2 can be, for example, —H, or a nitrogen protecting group selected from the group consisting of: 9-Fluorenylmethoxycarbonyl (Fmoc), Trityl (Trt), 4-Methoxytrityl (Mmt), 2-(3,5-Dimethoxyphenyl)propan-2-yloxycarbonyl (Ddz), 2-(p-Biphenylyl)-2-propyloxycarbonyl (Bpoc), 2-(4-Nitrophenylsulfonyl)ethoxycarbonyl (NSC), (1,1-Dioxobenzo[b]thiophene-2-yl)methyloxycarbonyl (Bsmoc), (1,1-Dioxonaphtho[1,2-b]thiophene-2-yl)methyloxycarbonyl (α-Nsmoc), 1-(4,4-dimethyl-2,6-dioxocyclohex-1-ylidene)-3-methylbutyl (ivDde), 2,-Di-tert-butyl-Fmoc (Fmoc*), 2-Fluoro-Fmoc (Fmoc(2F)), 2-Monoisooctyl-Fmoc (mio-Fmoc), 2,7-Diisooctyl-Fmoc (dio-Fmoc), 2-[Phenyl(methyl)sulfonio]ethyloxy carbonyl tetrafluoroborate (Pms), Ethanesulfonylethoxycarbonyl (Esc), 2-(4-Sulfophynylsulfonyl)ethoxy carbonyl (Sps), Tert-butyloxycarbonyl (Boc), Benzyloxycarbonyl (Z), Allyloxycarbonyl (Alloc), 2,2,2-Trichloroethyloxycarbonyl (Troc), p-Nitrobenzyloxycarbonyl (pNZ), Propargyloxycarbonyl (Poc), o-Nitrobenzenesulfonyl (oNBS), 2,4-Dinitrobenzenesulfonyl (dNBS), Benzothiazole-2-sulfonyl (Bts), o-Nitrobenzyloxycarbonyl (oNz), 4-Nitroveratryloxycarbonyl (NVCO), 2-(2-Nitrophenyl)propyloxycarbonyl (NPPOC), 2,(3,4-Methylethenedioxy-6-nitrophenyl)propyloxycarbonyl (MNPPOC), 9-(4-Bromophenyl)-9-fluorenyl (BrPhF), Azidomethoxycarbonyl (Azoc), Hexafluoroacetone (HFA), 2-Chlorobenzyloxycarbonyl (Cl-Z), 4-Methyltrityl (Mtt), Trifluoroacetyl (tfa), (Methylsulfonyl)ethoxycarbonyl (Msc), Phenyldisulphanylethyloxycarbonyl (Phdec), 2-Pyridyldisulphanylethyloxycarbonyl (Pydec), and o-Nitrobenzenesulfonyl (O-NBS).
  • Nitrogen protecting groups can be found, for example, in Isidro-Llobet, A., et al., “Amino Acid-Protecting Groups,” Chem. Rev. 2455-2504 (2009).
  • In the crystalline compound of Formula (I) or its crystalline salt, R2 can be, for example, a nitrogen protecting group selected from the group consisting of 9-Fluorenylmethoxycarbonyl (Fmoc), Trityl (Trt), 4-Methoxytrityl (Mmt), 2-(3,5-dimethoxyphenyl)propan-2-yloxycarbonyl (Ddz), 2-(p-biphenylyl)-2-propyloxycarbonyl (Bpoc), 2-(4-Nitrophenylsulfonyl)ethoxycarbonyl (NSC), 1,1-Dioxobenzo[b]thiophene-2-yl)methyloxycarbonyl (Bsmoc), 1-(4,4-dimethyl-2,6-dioxocyclohex-1-ylidene)-3-methylbutyl (ivDde), Tert-butyloxycarbonyl (Boc), Benzyloxycarbonyl (Z), Allyloxycarbonyl (Alloc), 2,2,2-Trichloroethyloxycarbonyl (Troc), p-Nitrobenzyloxycarbonyl (pNZ), o-Nitrobenzenesulfonyl (oNBS), 2,4-Dinitrobenzenesulfonyl (dNBS), o-Nitrobenzyloxycarbonyl (oNz), 4-Nitroveratryloxycarbonyl (NVCO), 2-(2-Nitrophenyl)propyloxycarbonyl (NPPOC), Hexafluoroacetone (HFA), 2-Chlorobenzyloxycarbonyl (Cl-Z), 4-Methyltrityl (Mtt), Trifluoroacetyl (tfa), (Methylsulfonyl)ethoxycarbonyl (Msc), and o-Nitrobenzenesulfonyl (O-NBS).
  • In the crystalline compound of Formula (I) or its crystalline salt, R2 can be a nitrogen protecting group selected from the group consisting of 9-Fluorenylmethoxycarbonyl (Fmoc), Trityl (Trt), 4-Methoxytrityl (Mmt), 2-(3,5-dimethoxyphenyl)propan-2-yloxycarbonyl (Ddz), 2-(p-biphenylyl)-2-propyloxycarbonyl (Bpoc), Tert-butyloxycarbonyl (Boc), Benzyloxycarbonyl (Z), Allyloxycarbonyl (Alloc), 2,2,2-Trichloroethyloxycarbonyl (Troc), o-Nitrobenzenesulfonyl (oNBS), Trityl (Trt), 4-Methyltrityl (Mtt), and o-Nitrobenzenesulfonyl (O-NBS).
  • In the crystalline compound of Formula (I) or its crystalline salt, R2 can be, for example, the nitrogen protecting group 9-Fluorenylmethoxycarbonyl (Fmoc).
  • R3
  • In the crystalline compound of Formula (I) or its crystalline salt, R3 can be, for example, —H or a protecting or activating group selected from the group consisting of: tert-Butyl (tBu), 2-Chlorotrityl (2-Cl-Trt), 2,4-Dimethoxybenzyl (DMB), Benzyl (Bn), 2-Phenylisopropyl (2-PhiPr), 5-Phenyl-3,4-ethylenedioxythenyl, 9-Fluorenylmethyl (Fm), 4-(N-[1-(4,4-dimethyl-2,6-dioxocyc lohexylidene)-3-methylbutyl]-amino)benzyl (Dmab), Methyl (Me), Ethyl (Et), Carbamoylmethyl (Cam), Allyl (Al), Phenacyl (Pac), p-Nitrobenzyl (pNB), 2-Trimethylsilylethyl (TMSE), (2-Phenyl-2-trimethylsilyl)ethyl (PTMSE), 2-(Trimethylsilyl)isopropyl (Tmsi), Trimethylsilyl (TMS), 2,2,2-Trichloroethyl (Tce), p-Hydroxyphenacyl (pHP), 4,5-Dimethoxy-2-nitrobenzyl (Dmnb), 1,1-Dimethylallyl (Dma), Pentaamine cobalt (III), Succinimide, p-Nitrophenyl, Pentafluorophenyl, and 2,4,5-trichlorophenyl.
  • In the crystalline compound of Formula (I) or its crystalline salt, R3 can be, for example —H.
  • n
  • In the crystalline compound of Formula (I) or its crystalline salt, n can range, for example, from 1-20, from 3-11, or from 3-6. n can be, for example 3 or 6 or 11. n can be 3. n can be 6. n can be 11.
  • *
  • In the crystalline compound of Formula (I) or its crystalline salt, the stereocenter * can be (R). In the crystalline compound of Formula (I) or its crystalline salt, the stereocenter * can be (S).
  • In one embodiment, in the crystalline compound of Formula (I) or it crystalline salt, R1 can be C1-C3 alkyl; R2 can be 9-Fluorenylmethoxycarbonyl (Fmoc); R3 can be selected from the group consisting of —H tert-Butyl (tBu), 2-Chlorotrityl (2-cl-Trt), 2,4-Dimethoxybenzyl (DMB), Benzyl (Bn), 2-Phenylisopropyl (2-PhiPr), 5-Phenyl-3,4-ethylenedioxythenyl, 9-Fluorenylmethyl (Fm), 4-(N-[1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl]-amino)benzyl (Dmab), Methyl (Me), Ethyl (Et), Carbamoylmethyl (Cam), Allyl (Al), Phenacyl (Pac), p-Nitrobenzyl (pNB), 2-Trimethylsilylethyl (TMSE), (2-Phenyl-2-trimethylsilyl)ethyl (PTMSE), 2-(Trimethylsilyl)isopropyl (Tmsi), Trimethylsilyl (TMS), 2,2,2-Trichloroethyl (Tce), p-Hydroxyphenacyl (pHP), 4,5-Dimethoxy-2-nitrobenzyl (Dmnb), 1,1-Dimethylallyl (Dma), Pentaamine cobalt (III), Succinimide, p-Nitrophenyl, Pentafluorophenyl, and 2,4,5-trichlorophenyl; n can be an integer ranging from 3 to 11; and the stereocenter * can be (R).
  • In one embodiment, in the crystalline compound of Formula (I) or it crystalline salt, R1 can be C1-C3 alkyl; R2 can be 9-Fluorenylmethoxycarbonyl (Fmoc); R3 can be selected from the group consisting of —H tert-Butyl (tBu), 2-Chlorotrityl (2-cl-Trt), 2,4-Dimethoxybenzyl (DMB), Benzyl (Bn), 2-Phenylisopropyl (2-PhiPr), 5-Phenyl-3,4-ethylenedioxythenyl, 9-Fluorenylmethyl (Fm), 4-(N-[1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl]-amino)benzyl (Dmab), Methyl (Me), Ethyl (Et), Carbamoylmethyl (Cam), Allyl (Al), Phenacyl (Pac), p-Nitrobenzyl (pNB), 2-Trimethylsilylethyl (TMSE), (2-Phenyl-2-trimethylsilyl)ethyl (PTMSE), 2-(Trimethylsilyl)isopropyl (Tmsi), Trimethylsilyl (TMS), 2,2,2-Trichloroethyl (Tce), p-Hydroxyphenacyl (pHP), 4,5-Dimethoxy-2-nitrobenzyl (Dmnb), 1,1-Dimethylallyl (Dma), Pentaamine cobalt (III), Succinimide, p-Nitrophenyl, Pentafluorophenyl, and 2,4,5-trichlorophenyl; n can be an integer ranging from 3 to 11; and the stereocenter * can be (S).
  • In one embodiment, in the crystalline compound of Formula (I) or its crystalline salt, R1 can be methyl, R2 can be 9-Fluorenylmethoxycarbonyl (Fmoc); R3 can be —H, n can be 3, 6, or 11, and the stereocenter * can be (R).
  • In one embodiment, in the crystalline compound of Formula (I) or its crystalline salt, R1 can be methyl, R2 can be 9-Fluorenylmethoxycarbonyl (Fmoc); R3 can be —H, n can be 3, 6, or 11, and the stereocenter * can be (S).
  • Chemical Purity
  • Herein, unless otherwise indicated, any compound, its salt, crystalline compound, or crystalline salt of a compound, can have a chemical purity. Chemical purity can be defined, for example, as the degree to which a substance is undiluted or unmixed with extraneous material, and can be typically expressed as a percentage. Any compound, salt thereof, crystalline compound, or crystalline salt of a compound herein can have, for example, a chemical purity ranging from about 90% to 100%. The chemical purity can be, for example, about 92% to 100%, about 94% to 100%, about 96% to 100%, about 98% to 100%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100%. The percentage can be, for example, based on the total weight of the compound, its salt, crystalline compound, or its salt. The percentage can be, for example, arrived at using HPLC. The percentage can be arrived at, for example, using NMR, for example proton NMR. The chemical purity can be arrived at, for example, using elemental analysis.
  • Enantiomeric Excess
  • Herein, unless otherwise indicated, any compound, salt thereof, crystalline compound, or crystalline salt of a compound, can have an enantiomeric excess. The enantiomeric excess can be, for example, from about 80% to 100%, from about 85% to 100%, from about 90% to 100%, from about 95% to 100%, from about 96% to 100%, from about 97% to 100%, from about 98% to 100%, from about 99% to 100%, about 95%, about 96%, about 97%, about 97.2%, about 98%, about 99%, or 100%. The enantiomeric excess can be, for example, greater than 95%, greater than 96%, greater than 97%, greater than 98%, or greater than 99%. Herein, unless otherwise indicated, enantiomeric excess can be calculated, for example, by the formula: enantiomeric excess (ee)=((P−S)/(P+S))×100%, where P and S represent the moles, respectively, of the predominant and subdominant enantiomer produced or present in a sample. For example, if the more moles of the (R) enantiomer are produced at than moles of the (S) enantiomer, moles of (R) enantiomer are designated as R, and moles of the (S) enantiomer are designated as S, then the enantiomeric excess formula becomes: ee (%)=((R−S)/(R+S))×100%. Herein, unless otherwise indicated, the amount (e.g., moles) or enantiomer produced can be determined, for example, by chiral HPLC, by chiral GC, or via a chiral NMR shift reagent using NMR spectroscopy.
  • Optical Purity
  • Herein, unless otherwise indicated, any compound, its salt, crystalline compound, or crystalline salt of a compound, can have an optical purity. The optical purity can be, for example, from about 80% to 100%, from about 85% to 100%, from about 90% to 100%, from about 95% to 100%, about 95%, about 96%, about 97%, about 98%, about 99%, or 100%. Herein, unless otherwise indicated, optical purity can be calculated using the formula: optical purity (%)=(Mobserved/Mmaximal)*100%, where [α]observed is the specific rotation of the sample, and [α]maximal is the specific rotation of the pure enantiomer. Herein, unless otherwise indicated, specific rotation can be defined as the observed angle of optical rotation, a, when plane-polarized light is passed through a sample with a path length of 1 decimeter and a sample concentration of 1 gram per 1 millilitre. The specific rotation can be obtained, for example, at 20° C. and at a wavelength of light of 589 nanometers (e.g., the sodium D line). Herein, unless otherwise indicated, the specific rotation can be obtained, for example, with a polarimeter. Herein, unless otherwise indicated, the solvent the sample is dissolved in can be any suitable solvent or solvent combination, for example, ethanol, methanol, chloroform, dichloromethane, carbon tetrachloride, water, DMSO, N,N-DMF, diethyl ether, tetrahydrofuran, hexane, pentane, acetone, or any combination thereof.
  • Diastereomeric Excess
  • Herein, unless otherwise indicated, the compounds, salts, crystalline compounds, or crystalline salts of compounds herein can be diastereomers. When this is so, the compounds, crystalline compounds, or crystalline salts of compounds herein can have a diastereomeric excess of, for example, from about 80% to 100%, from about 85% to 100%, from about 90% to 100%, from about 95% to 100%, about 95%, about 96%, about 97%, about 98%, about 99%, or 100%. Herein, unless otherwise indicated, the diasteromeric excess, for example, in a mixture of two diastereomers, can be calculated, for example, by the formula: diastereomeric excess de %=((D1−D2)/(D1+D2))*100%, wherein D1 represents, for example, the mole or percent weight of a first and most abundant diastereomer, and D2 represents for example, the mole or percent weight of a second and least abundant diastereomer, where mole percent is used consistently (e.g., alone) in the calculation, or where percent weight is used consistently (e.g., alone) in the calculation.
  • Converted Enantiomeric Excess or Optical Purity
  • Unless otherwise indicated, any compound, salt thereof, crystalline compound, or crystalline salt thereof, herein, that is a diastereomer, can be converted to an enantiomer or enantiomeric mixture having one stereocenter (e.g., * in Formula (I)) by, for example, removal of a nitrogen protecting group (e.g., removal of the nitrogen protecting group R2 in the crystalline compound of Formula (I) or its crystalline salt that, together with the stereocenter *, creates a diastereomer), and the resulting enantiomer or enantiomeric mixture can then have its enantiomeric excess or optical purity determined as described herein. The resulting enantiomeric excess or optical purity, in these circumstances, is termed a converted enantiomeric excess or converted optical purity. The converted enantiomeric excess can be, for example, from about 80% to 100%, from about 85% to 100%, from about 90% to 100%, from about 95% to 100%, from about 96% to 100%, from about 97% to 100%, from about 98% to 100%, from about 99% to 100%, about 95%, about 96%, about 97%, about 97.2%, about 98%, about 99%, or 100%. The converted enantiomeric excess can be, for example, greater than 95%, greater than 96%, greater than 97%, greater than 98%, or greater than 99%. The converted optical purity can be, for example, from about 80% to 100%, from about 85% to 100%, from about 90% to 100%, from about 95% to 100%, about 95%, about 96%, about 97%, about 98%, about 99%, or 100%. Thus, any optionally crystalline diastereomer or its optionally crystalline salt herein, unless otherwise indicated, can have a converted enantiomeric excess or converted optical purity.
  • Specifically Exemplified Crystalline Compounds and Crystalline Salts Thereof
  • Herein, unless otherwise indicated, the crystalline compound of Formula (I) or its crystalline salt can be a compound of Formula (IIa) or its crystalline salt:
  • Figure US20140128581A1-20140508-C00006
  • Herein, unless otherwise indicated, the crystalline compound of Formula (I) or its crystalline salt can be a compound of Formula (IIb) or its crystalline salt:
  • Figure US20140128581A1-20140508-C00007
  • Herein, unless otherwise indicated, the crystalline compound of Formula (I) or its crystalline salt can be a compound of Formula (IIIa) or its crystalline salt:
  • Figure US20140128581A1-20140508-C00008
  • Herein, unless otherwise indicated, the crystalline compound of Formula (I) or its crystalline salt can be a compound of Formula (IIIb) or its crystalline salt:
  • Figure US20140128581A1-20140508-C00009
  • Deuterated and Halogenated Compounds and their Salts
  • Also provided herein, unless otherwise indicated, are optionally crystalline compounds and their optionally crystalline salts of Formula (IV):
  • Figure US20140128581A1-20140508-C00010
  • wherein R2, R3, n, and * are the same as in the crystalline compound or its crystalline salt of Formula (I), each E is independently selected from the group consisting of deuterium and halogen, and R4 is selected from the group consisting of C1-C3 alkyl, C1-C3 deuteroalkyl and C1-C3 haloalkyl.
  • Deuterium
  • Herein, unless otherwise indicated, for any deuterated: compound, its salt, crystalline compound, or its crystalline salt; greater than 90%, greater than 92%, greater than 94%, greater than 96%, or greater than 98%, of the deuterated: compound, its salt, crystalline compound, or its crystalline salt; has a deuterium atom at each position designated as deuterium (D) in the deuterated: compound, its salt, the crystalline compound, or its crystalline salt.
  • Methods of Making
  • The compounds and their salts herein can be advantageously made by methods disclosed herein that result in at least one of the following advantages:
      • the compounds or their salts that are produced are crystalline;
      • the compounds and their salts (both of which can be crystalline) are advantageously produced in high yield;
      • the compounds and their salts (both of which can be crystalline) are advantageously produced in high chemical purities;
      • the compounds and their salts (both of which can be crystalline) are advantageously produced in high enantiomeric excess, optical purity, diastereomeric excess, high converted enantiomeric excess, or high converted optical purity; or
      • the compounds and their salts (both of which can be crystalline) are produced without chromatographic purification (e.g., without chromatography).
  • Unless otherwise indicated, the compounds, their salts, crystalline compounds, and their crystalline salts, herein can be produced using for example exemplary Scheme I (with modifications that would be readily apparent to a skilled artisan). Scheme I depicts formation of the crystalline N-Fmoc-(R)-α-methyl-α-aminodec-9-enoic acid (i.e., the crystalline compound of Formula (IIa)). Sequence I starts with Boc-D-proline (i.e., the compound of Formula (V)). It is understood that by starting with Boc-L-proline, compounds with the opposite stereochemistry of the compound of Formula (IIa) can be produced (e.g., the compound of Formula (IIb) can be produced). It is also understood that the stereochemisty of the amino acid used to form the metal complex (e.g., alanine used to form the metal complex of Formula (XI) in Scheme I) is not dispositive of the stereochemistry in the resulting crystalline compound (e.g., of Formula (IIa)) or its crystalline salt.
  • Figure US20140128581A1-20140508-C00011
  • In Scheme I, Boc-D-proline (Compound of Formula (V)) is first reacted with 2-aminobenzophenone (compound of Formula (VI)) to form the compound of Formula (VII). Next, the compound of Formula (VII) is deprotected to form the HCl salt of the compound of Formula (VIII). A skilled artisan would readily understand that the synthetic scheme contemplates use of acids other than HCl, including organic acids and inorganic acids, for example, nitric acid, phosphoric acid, sulfuric acid, boric acid, hydrofloric acid, hydrobromic acid, and perchloric acid.
  • The salt of the compound of Formula (VIII) is next reacted with benzyl bromide, and for example, a base, to form the compound of Formula (IX). A skilled artisan would readily understand that substituted benzyl halides could be employed in place of benzyl bromide. For example, the following benzyl halides, where X=Cl, Br, or I, could be employed:
  • Figure US20140128581A1-20140508-C00012
  • Representative benzyl halides are found in Belokon, Y. N., et al., “Halo-substituted (S)—N-(2-benzoylphenyl)-1-benzylpyrrolidine-2-carboxamides as new chiral auxiliaries for the asymmetric synthesis of (S)-α-amino acids,”Russian Chemical Bulletin, International Edition, 51(8): 1593-1599 (2002). Further and different benzyl halides could also be employed:
  • Figure US20140128581A1-20140508-C00013
  • These representative benzyl halides are found in Saghiyan, A. S., et al., “New chiral NiII complexes of Schiff's bases of glycine and alanine for efficient asymmetric synthesis of α-amino acids,” Tedrahedron: Asymmetry 17: 455-467 (2006).
  • Next, the compound of Formula (IX) is reacted with L-alanine and Ni(NO3)2 to form the metal complex of Formula (XI). The skilled artisan would understand that other amino acids other than alanine could be employed in Scheme I. For example, glycine; 2-aminobutanoic acid, 2-aminopentanoic acid, and valine, for example in their D or L forms, could be employed. The Ni(NO3)2 can be a hydrate, for example, a hexahydrate. The reaction can be run in an alcoholic solvent, for example, methanol. The reaction can be run at an elevated temperature, for example, from about 40° C. to about 60° C. The reaction can be run in the presence of a base, for example, a hydroxide, for example an inorganic hydroxide, for example, potassium hydroxide. Other hydroxides are contemplated, including sodium hydroxide, cesium hydroxide, lithium hydroxide, magnesium hydroxide, and ammonium hydroxide.
  • To increase purity of the final product from Scheme I, the metal complex of Formula (XI) can be crystallized one or more times from one or more solvents, for example a cyclic ether and a non-cyclic ether. In one embodiment, the solvent is tetrahydrofuran and methyl tert-butyl ether. In some cases the ratio of the cyclic ether to the non-cyclic ether is at most 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10. In other cases the ratio of the cyclic ether to the non-cyclic ether is at least 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10. For example, some cases the metal complex of Formula (XI) is crystallized from a mixture of tetrahydrofuran and methyl tert-butyl ether in ratio of at most 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10. In other cases the ratio of and tetrahydrofuran and methyl tert-butyl ether is at least 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10. In some cases the ratio of tetrahydrofuran and methyl tert-butyl ether is 1.5:10. The metal complex of Formula (XI) may also be crystallized with esters, for example with ethyl acetate or isopropyl acetate. The product or crystallized product of Formula (IX) can alternatively or additionally be crystallized or recrystallized from a solvent, for example an alcohol, for example isopropyl alcohol. Other alcohols are contemplated, including methanol, ethanol, n-propanol, a butanol, n-butanol, iso-butanol, sec-butanol, and tert-butanol.
  • The metal complex of Formula (XI) is then alkylated with 8-bromooct-1-ene to form the alkylated metal complex of Formula (XII). The skilled artisan would understand that other alkylating agents, including other halo alkyl olefins, could be used in place of 8-bromooct-1-ene. For example, alkylating agents of the Formula (XV) could be used:
  • Figure US20140128581A1-20140508-C00014
  • wherein X is Cl, Br, or I, and n is an integer from 1 to 20. For example, n can be from 3 to 11, from 3 to 6, or 3 or 6. Some or all of the hydrogen atoms present in the compound of Formula (XV) can be replaced with deuterium atoms or halogen atoms. The alkylation can be performed in one or more solvents, for example a polar aprotic solvent, for example N,N-dimethyl formamide (DMF). The alkylation can be performed, for example, at a temperature of less than 20° C., for example, from less than 20° C. to 5° C., from less than 20° C. to 10° C., or at about 10° C. The skilled artisan would also understand that when glycine is used to form the metal complex, two alkylations could be performed one after the other. For example, the first alkylation could be performed using a C1-C3 alkane with a leaving group such as a halogen (e.g., methyl bromide, ethyl bromide, n-propyl bromide), or a C1-C3 deuteroalkane with a leaving group such as a halogen (e.g., CD3Br, CD3CD2Br, CD3CD2CD2Br), or a C1-C3 haloalkane with a leaving group such as a more reactive halogen than the other halogens in the haloalkane (e.g., CF3Br, CF3CF2Br, CF3CF2CF2Br). Then, the second alkylation could be performed using the alkylating agent of Formula (XV). The order of the first and second alkylations can be reversed.
  • Purification of Formula (XII) may be achieved by crystallization one or more times from one or more solvents including cyclic and non-cyclic ethers, esters, hexanes and heptanes. For example crystallization may be achieved using a combination of ethyl acetate and hexanes, ethyl acetate and heptanes, isopropyl acetate and hexanes, isopropyl acetate and heptanes, methyl tertiary-butyl ether and hexanes, methyl tertiary-butyl ether and heptanes or isopropyl acetate and methyl tertiary-butyl ether.
  • The metal complex of Formula (XII) is then cleaved with an acid, for example HCl, using one or more solvents, for example an ether, for example a cyclic ether, for example tetrahydrofuran, to form the amino acid HCl salt of Formula (XIII). The skilled artisan would understand that other acids in addition to HCl are contemplated, for example organic or inorganic acids, for example, nitric acid, phosphoric acid, sulfuric acid, boric acid, hydrofloric acid, hydrobromic acid, or perchloric acid. The salt of Formula (XIII) may be further purified by crystallization one or more times with one or more solvents. The solvent may be any suitable solvent including tetrahydrofuran, methyl tertiary-butyl ether, ethyl acetate, isopropyl acetate, ethanol, methanol, isopropanol, acetonitrile, or a combination thereof. In one embodiment, the solvent is acetonitrile.
  • The amino acid salt of Formula (XIII) is then nitrogen protected with a nitrogen protecting group, in this case an Fmoc group, and the cyclohexylamine addition salt of the protected amino acid is formed, yielding the protected amino acid cyclohexylamine salt of Formula (XIV). Formation of the salt of Formula (XIV) can be achieved in any suitable solvent including acetonitrile, methyl tertiary-butyl ether, tetrahydrofuran or a combination thereof. In one embodiment, the solvent is methyl tertiary-butyl ether. A skilled artisan would understand that other amines, for example other cyclic amines, for example cyclopropylamine, cyclobutyl amine, cyclopentylamine, cycloheptylamine, and cyclooctylamine, are contemplated. One of skill in the art would also readily understand that other nitrogen protecting groups are contemplated, for example the nitrogen protecting groups for R2 in the crystalline compound of Formula (I) or its crystalline salt herein.
  • The protected amino acid cyclohexylamine salt of Formula (XIV) can then be crystallized from one or more ethers, for example, two ethers, for example a cyclic ether and a non cyclic ether, for example tetrahydrofuran and methyl tert-butyl ether.
  • The crystallized amino acid cyclohexylamine salt of Formula (XIV) is then treated with sulfuric acid, and subsequently crystallized to form the crystalline compound of Formula (IIa). The skilled artisan would understand that acids other than sulfuric acid are contemplated, for example organic or inorganic acids, for example, nitric acid, phosphoric acid, sulfuric acid, boric acid, hydrofloric acid, hydrobromic acid, or perchloric acid. The crystallization can be performed using one or more solvents, for example two solvents, for example an alkane and haloalkane, for example hexanes and chloroform. In some cases the ratio of the alkane to the haloalkane is at least 6:1, 5:1, 4:1, 3:1, 2:1, or 1:10. In some cases the ratio of the alkane to the haloalkane is at most 6:1, 5:1, 4:1, 3:1, 2:1, or 1:10. For example, the crystalline compound of Formula (IIa) may be obtained by crystallization from a mixture of hexanes and chloroform in the ratio of at least 6:1, 5:1, 4:1, 3:1, 2:1, or 1:1. The crystalised IIa may also obtained by crystallization from a mixture of hexanes and chloroform in the ratio of at most 6:1, 5:1, 4:1, 3:1, 2:1, or 1:1. In some cases the ratio of hexanes and chloroform is 3:1.
  • The crystallization can be performed at a temperature ranging from, for example, about −5° C. to about −20° C., about −10° C. to about −20° C., or about −15° C. to −20° C.
  • The skilled artisan would understand, for example, that the crystalline compound of Formula (IIa) could be further activated or protected at its carboxylic acid function with, for example, a protecting or activating group R3 of the crystalline compound of Formula (I) or its crystalline salt. Unless otherwise indicated, the compounds, their salts, crystalline compounds, and their crystalline salts, herein can be produced using exemplary Scheme II (with modifications that would be readily apparent to a skilled artisan). Scheme II depicts formation of the crystalline N-Fmoc-(S)-α-methyl-α-aminohept-6-enoic acid (i.e., the crystalline compound of Formula (IIIa)). Sequence II starts with Boc-L-Proline (i.e., the compound of Formula (Va)). It is understood that by starting with Boc-D-proline, compounds with the opposite stereochemistry of the compound of Formula (IIIa) can be produced (e.g., the compound of Formula (III) can be produced). It is also understood that the stereochemisty of the amino acid used to form the metal complex, and whose alpha carbon atom is subsequently alkylated by the haloolefin (e.g., alanine in Formula (XIa)) is not dispositive of the stereochemistry in the resulting crystalline compound (e.g., of Formula (III)) or its crystalline salt.
  • Figure US20140128581A1-20140508-C00015
  • In Scheme II, Boc-L-proline (Compound of Formula (Va)) is first reacted with 2-aminobenzophenone (compound of Formula (VI)) to form the compound of Formula (VIIa). Next, the compound of Formula (VIIa) is deprotected to form the HCl salt of the compound of Formula (VIIIa). A skilled artisan would readily understand that the synthetic scheme contemplates use of acids other than HCl, including organic acids and inorganic acids, for example, nitric acid, phosphoric acid, sulfuric acid, boric acid, hydrofloric acid, hydrobromic acid, and perchloric acid.
  • The salt of the compound of Formula (VIIIa) is next reacted with benzyl bromide, and for example a base, to form the compound of Formula (IXa). A skilled artisan would readily understand that substituted benzyl halides could be employed in place of benzyl bromide. For example, the following benzyl halides, where X=Cl, Br, or I, could be employed:
  • Figure US20140128581A1-20140508-C00016
  • Representative benzyl halides are found in Belokon, Y. N., et al., “Halo-substituted (S)—N-(2 benzoylphenyl)-1-benzylpyrrolidine-2-carboxamides as new chiral auxiliaries for the asymmetric synthesis of (S)-α-amino acids,”Russian Chemical Bulletin, International Edition, 51(8): 1593-1599 (2002). Further and different benzyl halides could also be employed:
  • Figure US20140128581A1-20140508-C00017
  • These representative benzyl halides are found in Saghiyan, A. S., et al., “New chiral NiII complexes of Schiff's bases of glycine and alanine for efficient asymmetric synthesis of α-amino acids,” Tedrahedron: Asymmetry 17: 455-467 (2006).
  • Next, the compound of Formula (IXa) is reacted with L-alanine and Ni(NO3)2 to form the metal complex of Formula (XIa). The skilled artisan would understand that other amino acids other than alanine could be employed in Scheme II. For example, glycine; 2-aminobutanoic acid, 2-aminopentanoic acid, and valine could be employed, for example in their D or L forms. The Ni(NO3)2 can be a hydrate, for example, a hexahydrate. The reaction can be run in an alcoholic solvent, for example, methanol. The reaction can be run at an elevated temperature, for example, from about 40° C. to about 60° C. The reaction can be run in the presence of a base, for example, a hydroxide, for example an inorganic hydroxide, for example, potassium hydroxide. Other hydroxides are contemplated, including sodium hydroxide, lithium hydroxide, cesium hydroxide, and magnesium hydroxide.
  • To increase purity of the final product from Scheme II, the metal complex of Formula (XIa) can be crystallized one or more times from one or more solvents, for example a cyclic ether and a non-cyclic ether, for example tetrahydrofuran and methyl tert-butyl ether. In some cases the ratio of the cyclic ether to the non-cyclic ether is at most 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10. In other cases the ratio of the cyclic ether to the non-cyclic ether is at least 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10. For example, some cases the metal complex of Formula (XIa) is crystallized from a mixture of tetrahydrofuran and methyl tert-butyl ether in ratio of at most 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10. In other cases the ratio of and tetrahydrofuran and methyl tert-butyl ether is at least 0.5:10, 1.0:10, 1.5:10, 2.0:10, 2.5:10, 3.0:10, 3.5:10, 4.0:10, 4.5:10 or 5:10. In some cases the ratio of tetrahydrofuran and methyl tert-butyl ether is 1.5:10. The product or crystallized product of Formula (IXa) can be crystallized or recrystallized from a solvent, for example an alcohol, for example isopropyl alcohol. Other alcohols are contemplated, including methanol, ethanol, n-propanol, a butanol, n-butanol, iso-butanol, sec-butanol, and t-butanol. Other solvents suitable for crystallization or recrystallization of Formula (XIa) include esters, for example ethyl acetate or isopropyl acetate.
  • The metal complex of Formula (XIa) is then alkylated with 5-bromopent-1-ene to form the alkylated metal complex of Formula (XIIa). The skilled artisan would understand that other alkylating agents, including other halo alkyl olefins, could be used in place of 5-bromopent-1-ene. For example, alkylating agents of the Formula (XV) could be used:
  • Figure US20140128581A1-20140508-C00018
  • wherein X is Cl, Br, or I, and n is an integer from Ito 20. For example, n can be from 3 to 11, from 3 to 6, or 3 or 6. Some or all of the hydrogen atoms present in the compound of Formula (XV) can be replaced with deuterium atoms or halogen atoms. The alkylation can be performed in one or more solvents, for example a polar aprotic solvent, for example N,N-dimethyl formamide (DMF). The alkylation can be performed, for example, at a temperature of less than 20° C., for example, from less than 20° C. to 5° C., from less than 20° C. to 10° C., or at about 10° C. The skilled artisan would also understand that when glycine is used to form the metal complex, two alkylations could be performed one after the other. For example, the first alkylation could be performed using a C1-C3 alkane with a leaving group such as a halogen (e.g., methyl bromide, ethyl bromide, n-propyl bromide), or a C1-C3 deuteroalkane with a leaving group such as a halogen (e.g., CD3Br, CD3CD2Br, CD3CD2CD2Br), or a C1-C3 haloalkane with a leaving group such as a more reactive halogen than the other halogens in the haloalkane (e.g., CF3Br, CF3CF2Br, CF3CF2CF2Br). Then, the second alkylation could be performed using the alkylating agent of Formula (XV). The order of the first and second alkylations can be reversed.
  • Purification of Formula (XIIa) may be achieved by crystallization one or more times from one or more solvents including cyclic and non-cyclic ethers, esters, hexanes and heptanes. For example crystallization may be achieved by using a combination of ethyl acetate and hexanes, ethyl acetate and heptanes, isopropyl acetate and hexanes, isopropyl acetate and heptanes, methyl tertiary-butyl ether and hexanes, methyl tertiary-butyl ether and heptanes or isopropyl acetate and methyl tertiary-butyl ether.
  • The metal complex of Formula (XIIa) is then cleaved with an acid, for example HCl, using one or more solvents, for example an ether, for example a cyclic ether, for example tetrahydrofuran, to form the amino acid HCl salt of Formula (XIIIa). The skilled artisan would understand that other acids in addition to HCl are contemplated, for example organic or inorganic acids, for example, nitric acid, phosphoric acid, sulfuric acid, boric acid, hydrofloric acid, hydrobromic acid, or perchloric acid.
  • The salt of Formula (XIIIa) may be further purified by crystallization one or more times with one or more solvents. The solvent may be any suitable solvent including tetrahydrofuran, methyl tertiary-butyl ether, ethyl acetate, isopropyl acetate, ethanol, methanol, isopropanol, acetonitrile, or a combination thereof. In one embodiment, the solvent is acetonitrile.
  • The amino acid salt of Formula (XIIIa) is then nitrogen protected with a nitrogen protecting group, in this case an Fmoc group, yielding the protected amino acid of Formula (XIVa). In some embodiments, the compound of Formula (XIVa) is taken on to the crystallization step as is. In other embodiments, the compound of Formula (XIVa) is converted to a salt prior to crystallization. Formation of the salt of Formula (XIVa) may be achieved in any suitable solvent including acetonitrile, methyl tertiary-butyl ether, tetrahydrofuran or a combination thereof. One of skill in the art would also readily understand that other nitrogen protecting groups are contemplated, for example the nitrogen protecting groups for R2 in the crystalline compound of Formula (I) or its crystalline salt herein. For example, a protected amino acid cyclohexylamine salt of Formula (XIVa) can then be crystallized from one or more ethers, for example, two ethers, for example a cyclic ether and a non cyclic ether, for example tetrahydrofuran and methyl tert-butyl ether.
  • The protected amino acid cyclohexylamine salt of Formula (XIVa) can then be crystallized to form the crystalline compound of Formula (IIIa).
  • The crystallization can be performed using one or more solvents, for example two solvents, for example an alkane and haloalkane, for example hexanes and chloroform. In some cases the ratio of the alkane to the haloalkane is at least 6:1, 5:1, 4:1, 3:1, 2:1, or 1:10. In some cases the ratio of the alkane to the haloalkane is at most 6:1, 5:1, 4:1, 3:1, 2:1, or 1:10. For example, the crystalline compound of Formula (IIIa) may be obtained by crystallization from a mixture of hexanes and chloroform in the ratio of at least 6:1, 5:1, 4:1, 3:1, 2:1, or 1:1. The crystalised IIIa may also obtained by crystallization from a mixture of hexanes and chloroform in the ratio of at most 6:1, 5:1, 4:1, 3:1, 2:1, or 1:1. In some cases the ratio of hexanes and chloroform is 2:1.
  • The crystallization can be performed at a temperature ranging from, for example, about −5° C. to about −20° C., about −10° C. to about −20° C., or about −15° C. to −20° C. Herein, unless otherwise indicated, any compound or its salt may be crystalline. Herein, unless otherwise indicated, any compound or its salt may be crystalline at a temperature, for example, of about 0° C. or less, about −5° C. or less, about −10° C. or less, about −15° C. or less, about −20° C. or less, about −5° C., about −6° C., about −7° C., about −8° C., about −9° C., about −10° C., about −11° C., about −12° C., about −13° C., about −14° C., about −15° C., about −16° C., about −17° C., about −18° C., about −19° C., or about −20° C.
  • The skilled artisan would understand, for example, that the crystalline compound of Formula (IIIa) could be further activated or protected at its carboxylic acid function with, for example, a protecting or activating group R3 of the crystalline compound of Formula (I) or its crystalline salt.
  • Stapled and Stitched Polypeptides
  • The crystalline compounds and their crystalline salts of Formula (I), including the crystalline compounds and their crystalline salts of Formulae (IIa), (IIb), (IIIa) and (IIIb), as well as the optionally crystalline compounds and their optionally crystalline salts of Formula (IV), can be used to synthesize peptides, polypeptides, and crosslinked polypeptides that are useful for treating and preventing diseases.
  • The crosslinked polypeptides can contain secondary structures such as a helix, for example, an alpha helix. The crosslinker can stablize the secondary structures relative to an otherwise identical but uncrosslinked polypeptide. And the crosslinker can be formed by, for example, joining the terminal alkene side chains of, for example, two crystalline alkene α, α-disubstituted amino acids or their crystalline salts herein that are incorporated into a polypeptide through, for example, a metal catalyzed olefin metathesis reaction (e.g., forming a stapled peptide). This process is depicted in Scheme III, below:
  • Figure US20140128581A1-20140508-C00019
  • Examples of stapled polypeptides are found, inter alia, for example, in International Application No. PCT/US2004/038403.
  • The crystalline compounds and their crystalline salts of Formula (I), including the crystalline compounds and their crystalline salts of Formulae (IIa), (IIb), (IIIa) and (IIIb), as well as the optionally crystalline compounds and their optionally crystalline salts of Formula (IV), can be used to synthesize peptides, polypeptides, and stitched polypeptides that are useful for treating and preventing diseases.
  • For example, two of the crystalline compounds and their crystalline salts of Formula (I), can be incorporated into a polypeptide backbone along with an α, α-disubstituted amino acid having terminal olefins on each of its side chains, for example the compound of Formula (XVI):
  • Figure US20140128581A1-20140508-C00020
  • as shown in scheme IV. Metal catalyzed metathesis reaction of the olefins yields a stitched peptide.
  • Figure US20140128581A1-20140508-C00021
  • Examples of stitched polypeptides are found, for example, in International Application Publication No. WO2008/121767.
  • Methods to effect formation of peptidomimetic macrocycles which are known in the art can be employed. For example, the preparation of peptidomimetic macrocycles are described in Schafmeister et al., J. Am. Chem. Soc. 122:5891-5892 (2000); Schafineister & Verdine, J. Am. Chem. Soc. 122:5891 (2005); Walensky et al., Science 305:1466-1470 (2004); U.S. Pat. No. 7,192,713 and International Pat. App. Pub. No. WO 2008/121767.
  • Herein, unless otherwise indicated, the term “peptide synthesis” encompasses coupling of two or more amino acids with the aid of a coupling reagent. Peptide synthesis may be performed in “liquid” or “solution” phase where the coupling of the amino acids is performed in a solvent system. Peptide synthesis may also, or alternatively, be performed on “solid phase” where an amino acid is attached to a polymeric or solid support by a covalent bond at the N- or C-terminus of an amino acid. Peptides can be made, for example, by chemical synthesis methods, such as those described in Fields et al., Chapter 3 in Synthetic Peptides: A User's Guide, ed. Grant, W. H. Freeman & Co., New York, N.Y., 1992, p. 77,; and Goodman, M., et al., Houben-Weyl Methods in Organic Chemistry: Synthesis of Peptides and Peptidomimetics, Thieme Publishers, Volumes 1-5, (1994). For example, peptides can be synthesized using automated Merrifield techniques of solid phase synthesis with the amino groups of the amino acids employed in the synthesis protected, for example by t-Boc or Fmoc protecting groups. An automated peptide synthesizer (e.g., Applied Biosystems (Foster City, Calif.), Model 430A, 431, or 433) can be employed in making peptides.
  • Herein unless otherwise indicated, peptidomimetic precursors and peptidomimetic macrocycles and their salts described herein can be produced using solid phase peptide synthesis (SPPS), where for example, a C-terminal amino acid is attached to a cross-linked polystyrene resin via an acid or base labile bond with a linker. The resin can be, for example, insoluble in the solvents used for synthesis, making it relatively simple and fast to wash away excess reagents and by-products. The N-terminus of each amino acid added to the growing peptide chain can be protected, for example, with an Fmoc group, which is stable in acid, but removable by base. Side chain functional groups can be protected, as necessary or desirable, for example, with base stable, acid labile groups.
  • Herein, unless otherwise indicated, the peptidomimetic precursors can be made, for example, in a high-throughput, combinatorial fashion using, for example, a high-throughput polychannel combinatorial synthesizer (e.g., Thuramed TETRAS multichannel peptide synthesizer from CreoSalus, Louisville, Ky. or Model Apex 396 multichannel peptide synthesizer from AAPPTEC, Inc., Louisville, Ky.).
  • Herein, unless otherwise indicated, solution peptide synthesis can be performed in a manner wherein reagents are fully or partially dissolved in, for example, an appropriate solvent, for example, a polar aprotic solvent. In a representative case employing, for example, a solid crystalline N-terminally protected olefinic amino acid with a removable protecting group (e.g., t-Butyloxycarbonyl, Benzyloxycarbonyl, Fluorenylmethoxycarbonyl) and a C-protected amino acid with a selectively removable ester (e.g., methyl, benzyl, t-butyl), the amino acids can be fully or partially dissolved in a solvent and an activating agent is added to accomplish peptide bond formation between the amino acids. Solution peptide synthesis can also utilize first formation of active esters of N-protected olefinic amino acids (e.g., N-hydroxysuccinamide, p-nitrophenyl, 2,4,6-trichlorophenyl, pentafluorophenyl) and then subsequent reaction of the activated amino acid with an unprotected or C-protected amino acid. The active esters of olefinic amino acids can be prepared, for example, by reacting a solid N-protected olefinic amino acid with an appropriate alcohol with help of the condensing agent (e.g., dicyclohexylcarbodiimide). These same procedures can also be used, for example, when one or both of the amino acids to be reacted are part of, and incorporated into, respectively, for example, one or two peptides.
  • Formation of C-terminally protected olefinic amino acids can easily be facilitated by reacting dry solid olefinic amino acid(s) with an appropriate alcohol (e.g., methyl, ethyl, benzyl) under, for example, anhydrous conditions. Formation of a peptide where olefinic amino acid is located in the C-terminal position can accomplished, for example, in the similar way. Solution methods of peptide preparation can be easily adapted to process scale. The starting materials and reagents used herein in preparing any compound herein and as above-and-below disclosed, unless otherwise indicated, for example, can be available from commercial sources such as Aldrich, Sigma or Bachem, or can be prepared by methods known to those skilled in the art following procedures set forth, for example, in references such as: Fieser and Fieser's Reagents for Org. Syn. Vol. 1-17, Organic Reactions Vol. 1-40, March's Advanced Organic Synthesis, Larock's Comprehensive Organic Transformations, Bodansky and Bodansky's The Practice of Peptide Synthesis, Greene's Protective Groups in Organic Synthesis, Wei, Q., et al., Tetrahedron 56: 2577-2582 (2000), Belokon, Y. N., et al., Tetrahedron: Asymmetry 9: 4249-4252 (1998), Belokon, Y., Pure & App. Chem. 64(12): 1917-1924 (1992), Ueki, H., et al., J. Org. Chem. 68: 7104-7107 (2003).
  • These schemes herein are illustrative of some methods by which compounds herein and their salts (which can be crystalline) can be synthesized, and various modifications to these schemes can be made and will be suggested to one skilled in the art having referred to this disclosure.
  • The starting materials and intermediates of the reactions of any embodiment herein, herein and as-above disclosed, unless otherwise indicated, may be isolated and purified if desired using conventional techniques, including, but not limited to filtration, distillation, crystallization, chromatogram, flash chromotography, HPLC, MPLC, Chromatotron®, ion exchange chromatography, crystallization with Mosher acids or Mosher esters, and the like. Such materials may be characterized using conventional means, including physical constructs and spectral data, for example proton NMR, carbon NMR, IR spectroscopy, polarimetry, atomic absorption, elemental analysis, UV spectroscopy, FTIR spectroscopy, and the like. In any embodiment here and as-above described, unless otherwise indicated, chromatography can be excluded in making any of the compounds or their salts.
  • Unless specified to the contrary, the reactions described herein can take place at, for example, from about 0.001 to about 100 atmospheres (atm), for example, about 0.001 atm, about 0.01 atm, about 0.1 atm, about 1 atm, about 2 atm, about 3 atm, about 4 atm, about 5 atm, about 10 atm, about 20 atm, about 50 atm, or about 100 atm.
  • Reactions in any embodiment herein, unless otherwise indicated, can be run, unless otherwise specified, for example, open to the atmosphere, or under an inert gas atmosphere such as, for example, nitrogen or argon.
  • Reactions in any embodiment herein, unless otherwise indicated, can be run, unless otherwise specified, for example, at temperatures from about −78° C. to about 150° C., for example from about −78° C., about −50° C., about −20° C., about 0° C., about 10° C., about 20° C., about 23° C., about 25° C., about 27° C., about 30° C., about 40° C., about 50° C., about 100° C., about 125° C., about 150° C., at about ambient temperature, or at about room temperature.
  • Reactions herein, unless otherwise indicated, can have a yield, unless otherwise explicitly stated, based on the theoretical yield, for example, ranging from about 1% to about 99%. The yield can be, for example, about 99%, about 98%, about 97%, about 96%, about 95%, about 90%, about 85%, about 80%, about 75%, about 70%, about 65%, about 60%, about 55%, about 50%, about 45%, about 40%, about 35%, about 30%, about 25%, about 20%, about 15%, about 10%, or about 5%.
  • Reactions herein, unless otherwise indicated, can be run, unless otherwise specified, for example, for a time ranging from about 0.1 to about 96 hours, e.g., for about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 13 hours, about 14 hours, about 15 hours, about 16 hours, about 17 hours, about 18 hours, about 19 hours, about 20 hours, about 21 hours, about 22 hours, about 23 hours, about 24 hours, about 48 hours, about 72 hours, or about 96 hours,
  • Selective Uses of Crosslinked Peptidomimetic Macrocycles (Stitched and Stapled Peptides)
  • Crosslinked peptidomimetic macrocycles (stitched or stapled peptides), made with for example at least one of the crystalline compounds and their crystalline salts of Formula (I), including the crystalline compounds and their salts of Formulae (IIa), (IIb), (IIIa) and (IIIb), as well as the optionally crystalline compounds and their optionally crystalline salts of Formula (IV), can be used to treat or prevent diseases. For example, the crosslinked peptidomimetic macrocycles (stitched or stapled peptides) can be used to treat or prevent cancers. Selected examples of cancers include, for example, fibrosarcoma, myosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, gastric cancer, esophageal cancer, rectal cancer, pancreatic cancer, ovarian cancer, prostate cancer, uterine cancer, cancer of the head and neck, skin cancer, brain cancer, squamous cell carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular cancer, small cell lung carcinoma, non-small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, retinoblastoma, leukemia, lymphoma, or Kaposi sarcoma.
  • Diseases which can be treated by stitched or stapled peptides can be found, for example, in International Application No. PCT/US2004/038403 (“the '403 application”) and International Application Publication No. WO2008/121767 (“the '767 publication”).
  • While inventive embodiments have been shown and described herein, such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the inventive disclosure herein. The following Examples are illustrative and should not be construed as limiting.
  • EXAMPLES Example 1 Preparation of crystalline N-Fmoc-(R)-α-methyl-α-aminodec-9-enoic acid Example 1a Preparation of (R)-2-[N—(N′-Boc-prolyl)amino]benzophenone
  • Figure US20140128581A1-20140508-C00022
  • Tetrahydrofuran and 9.6 kg (1.0 equivs.) of Boc-D-proline (V) were added to a reactor and cooled to −5° C. 5.3 kg (1.15 equivs.) of N-methylmorpholine were charged followed by a slow addition of 6.1 kg (1.0 equivs.) of isobutyl chloroformate in tetrahydrofuran while maintaining the internal temperature at <5° C. The mixture was allowed to agitate at 20-25° C. for 45-60 minutes and then was analyzed by TLC for completion. A solution of 8.2 kg (0.9 equvs.) of 2-aminobenzophenone/tetrahydrofuran was charged and the mixture was allowed to agitate at 20-25° C. until the reaction is deemed complete. The mixture was concentrated to ½ volume and isopropyl acetate was charged. The organic product layer was then washed with a 5% sodium bicarbonate solution, water was charged, then the pH was adjusted to 2.0-2.5 with 25% sulfuric acid. The layers were split and the organic product layer was washed again with water. The organic product solution was then concentrated and crystallized from isopropyl acetate and washed with methyl tert-butyl ether. Product (VII) was isolated and dried under heat and vacuum. Yield: 12 kg, 66.7%.
  • Example 1b Preparation of D-Proline-2-Aminobenzophenone amide
  • Figure US20140128581A1-20140508-C00023
  • 12.0 kg (1.0 equivs.) of Boc-D-proline-2-aminobenzophenone (VII) amide was dissolved into acetonitrile.
  • 2.2 kg (2.0 equivs.) of hydrogen chloride gas was then charged/bubbled into the solution. The resulting mixture was then allowed to agitate at 20-25° C. until the reaction was complete. Methyl tert-butyl ether was added and the solid product was isolated out of the reaction solution and washed with additional methyl tert-butyl ether. The product (VIII) was dried under heat and vacuum. Yield: 9.1 kg, 100%.
  • Example 1c Preparation of (R)-2-[N—(N′-benzylprolyl)amino]benzophenone (D-BPB)
  • Figure US20140128581A1-20140508-C00024
  • 9.1 kg (1.0 equivs.) of D-proline-2-aminobenzophenone amide.HCl (VIII) was dissolved into tetrahydrofuran and water. 8.1 kg (2.4 equivs.) of triethylamine was then charged, followed by a slow addition of 7.9 kg (1.4 equivs.) of benzyl bromide. The mixture was then allowed to agitate at 20-25° C. until the reaction was complete. Methyl tert-butyl ether and water were added and the resulting solution was pH adjusted to 2.0-2.5 with a 1N hydrochloric acid solution. The mixture was concentrated to remove all the tetrahydrofuran. The product slurry was then isolated and washed with methyl tert-butyl ether. The product (IX) was dried under heat and vacuum Yield: 10.5 kg, 82.7%.
  • Example 1d Preparation of (R)-Ala-Ni-BPB
  • Figure US20140128581A1-20140508-C00025
  • 10.5 kg (1.0 equivs.) of D-BPB (IX), 14.1 kg (1.78 equivs.) nickel (II) nitrate hexahydrate, 4.9 kg (2.0 equivs) of L-alanine, and methanol were charged to a reactor. The mixture was heated to 40° C. and a solution of 12.2 kg (8.0 equivs.) of potassium hydroxide/methanol was slowly added while maintaining the internal temperature of <50° C. The reaction mixture was then heated up to 60° C. and allowed to agitate at temperature until the reaction was complete. The mixture was then cooled to 20-25° C. and 8.2 kg (5.0 equivs.) of acetic acid was slowly charged while maintaining an internal temperature of <35° C. The reaction solution was concentrated to a solid. Tetrahydrofuran and isopropyl acetate were then added to dissolve the solid(s) and the organic product layer was washed 2× with water. The solution was then concentrated again and material was subsequently crystallized out of tetrahydrofuran and methyl tert-butyl ether. The product was isolated, rinsed with additional methyl tert-butyl ether and analyzed for purity. To improve purity the product (XI) was recrystallized out of isopropyl alcohol and then isolated, and dried under heat and vacuum. Yield: 6.8 kg, 48.6%.
  • Recrystallization Procedure
  • THF was added to the crude product (15 mL per 10 g of starting material (D-BPB)) and the resulting mixture was heated to 50° C. The mixture was maintained at 50° C. for 1 h, then methyl tertiary-butyl ether was added (50 mL per 10 g of starting material (D-BPB)). The mixture was maintained at 50° C. for additional 1 h after which it was cooled to 35° C. The mixture was filtered and the resulting solid was washed with methyl tertiary-butyl ether (20 mL per 10 g of starting material (D-BPB)) to obtain the crystalline product XI.
  • Alternate Recrystallization Procedure
  • Isopropyl acetate was added to the crude product (40 mL per 4 g of starting material (D-BPB)) and the resulting mixture was maintained at room temperature for 30 min. The mixture was then filtered to obtain the crystalline product XI.
  • Example 1e Preparation of R8-Ni-BPB
  • Figure US20140128581A1-20140508-C00026
  • 6.8 kg (1.0 equivs.) of (R)-Ala-Ni-BPB (XI) was charged to a reactor and dissolved up into dimethylformamide and cooled to 10° C. 1.4 kg (2.5 equivs.) of sodium hydroxide (powder) was then charged to the same reactor and the mixture was sparged with nitrogen and agitated until a solution formed at 10° C. 5.2 kg (2.0 equivs.) of 8-bromo-1-octene was charged to the reactor while maintaining an internal temperature of <20° C. The mixture was then allowed to agitate at 20-25° C. until the reaction was complete. Once the reaction was complete, the solution was cooled to 10° C. and 0.5 kg (0.6 equivs.) of acetic acid was charged maintaining the internal temperature <25° C. Water was then charged followed by methyl tert-butyl ether and the organic layer was washed. The organic layer was then washed 2 more times with water and then concentrated. The product oil was then co-stripped with methylene chloride and dissolved up in additional methylene chloride. The product (XII) solution was taken on into the next processing step.
  • Example 1f Preparation of (R)-2-Amino-2-methyl-dec-9-enoic acid
  • Figure US20140128581A1-20140508-C00027
  • The R8-Ni-BPB (XII)/DCM solution was charged to a 50-L chem-glass reactor and stripped to an oil.Tetrahydrofuran was then added and the mixture was agitated at 20-25° C. until a solution formed. 7.8 kg (5.0 equvs.) of 32% hydrochloric acid was charged slowly while maintaining an internal temperature of <30° C. The mixture was then allowed to agitate for 6-8 hours at ambient temperature. The mixture was then concentrated to remove tetrahydrofuran to yield a slurry. Additional water was added and the slurry was agitated at ambient temperature for 1-2 hours. The solid BPB salts were isolated by filtration and rinsed with additional water followed by methyl tert-butyl ether. The product filtrates were then re-charged to the reactor yielding a tri-phased solution. The lower-most layer was split from the upper two layers. The combined two organic layers were then washed 3× with water and concentrated to an oil. Acetonitrile was then added and the mixture was warmed to 70° C. for 30 minutes. The mixture was then cooled to 25-30° C. and the solid product was isolated. The solid filter-cake was washed with acetonitrile and methyl tert-butyl ether, then analyzed for purity. The product was then re-slurried out of additional acetonitrile and washed with acetonitrile and methyl tert-butyl ether. The material (XIII) was isolated and dried under heat and vacuum. Yield: 1.55 kg, 48%.
  • Recrystallization Procedure
  • Acetonitrile (23 mL per 10 g of starting material (oil of (R)-Ala-Ni-BPB (XI))) was added to the crude product and the resulting mixture was heated to 70° C. for 30 min after which it was cooled to 20° C. The mixture was filtered and the resulting solid was washed with acetonitrile (5 mL) and methyl tertiary-butyl ether (8.5 mL) to obtain the crystalline product XIII.
  • Alternate Recrystallization Procedure to Prepare XIII-II
  • Acetonitrile (30 mL per 10 g of starting material (oil of (R)-Ala-Ni-BPB (XI))) was added to the crude product and the resulting mixture was heated at 60° C. for 30 min followed by cooling to 30° C. The mixture was then filtered and washed with 5 mL acetonitrile to obtain the crystalline product XIII.
  • Alternate Recrystallization Procedure to Prepare XIII-II
  • Acetonitrile (23 mL per 10 g of starting material (oil of (R)-Ala-Ni-BPB (XI))) was added to the crude product and the resulting mixture was heated at 40° C. for 30 min followed by cooling to room temperature. The mixture was then filtered and washed twice with 5 mL acetonitrile to obtain the crystalline product XIII.
  • Example 1g Preparation of Crystalline N-Fmoc-(R)-α-methyl-α-aminodec-9-enoic acid
  • Figure US20140128581A1-20140508-C00028
  • 1.55 kg (1.0 equiv.) of 2-amino-2-methyl-dec-9-enoic acid.HCl (XIII) was suspended in water and polished filtered to remove trace amounts of D-BPB.HCl from the solution. Methyl tert-butyl ether was added and the aqueous product layer was extracted once with methyl tert-butyl ether. The aqueous product layer was re-charged and tetrahydrofuran was added. A 20% aqueous sodium carbonate solution (2.75 equiv.) was charged to the mixture followed by Fmoc-OSu (0.89 equiv.). The mixture was allowed to react at 20-25° C. while maintaining the pH between 8.5-9.0 with additional amounts of the 20% sodium carbonate solution until the reaction was complete. The mixture was pH adjusted down to pH 2.0-2.5 with conc. hydrochloric acid. Tetrahydrofuran was distilled off and methyl tert-butyl ether was charged. The layers were separated and the organic layer was washed 3 more times with additional water. The organic layer was then concentrated under vacuum and co-stripped with methyl tert-butyl ether. The resulting crude oil was re-dissolved in methyl tert-butyl ether and cyclohexylamine (1.10 equiv.) was added slowly to obtain a pH range of 8.5-9.0. The slurry was agitated at ambient temperature (20-25° C.) for 3 hours and the solid product salt (XIV) was isolated by filtration. The solids were rinsed twice with additional methyl tert-butyl ether and the solid wetcake was recharged to a clean reactor. The wetcake was recrystallized from tetrahydrofuran and methyl tert-butyl ether to improve the purity. The solid salt was suspended in methyl tert-butyl ether and water and the pH adjusted to 2.0-2.5 with 25% sulfuric acid. The organic product layer was washed with water until all of the cyclohexylamine was removed. The organic product layer was concentrated and co-stripped with hexanes to a loose oil. The product (IIa) was then crystallized out of chloroform and hexanes and dried at <0° C. under a 1.0 cfm nitrogen sweep. Yield: 1.12 kg, 41.5%
  • Recrystallization Procedure
  • Methyl tertiary-butyl ether (800 mL per 36 g of starting material XIII) was added to the crude product and the pH of the resulting mixture was adjusted to 8-9 using CHA at 20° C. The mixture was mixed at 20° C. and after 1 h crystals started forming. Additional methyl tertiary-butyl ether was added (200 mL) and the resulting slurry was mixed for 18 h. The mixture was filtered and the resulting solid was washed with twice methyl tertiary-butyl ether (200 mL and 8.5 mL) to obtain the crystalline product XIII. The product was analyzed for chiral purity, and if the results were less than 95% Fmoc-R8 vs. Fmoc-S8 then crystallization was performed to upgrade the chiral purity by dissolving dry FmocR/S (50 g) in THF (50 mL). Once FmocR/S was dissolved, methyl tertiary-butyl ether was added (900 mL) and the mixture was mixed at 20° C. for 18 h. The mixture was then filtered and washed twice with methyl tertiary-butyl ether (100 mL each). The chiral purity of the resulting crystalline product XIV was about 97.8%
  • Alternate Recrystallization Procedure for XIV-I
  • Methyl tertiary-butyl ether (1500 mL per 47 g of starting material XIII) was added to the crude product and the pH of the resulting mixture was adjusted to 8-9 using CHA at 20° C. The mixture was mixed at this temperature for 3 h after which it was filtered and the resulting solid was washed with methyl tertiary-butyl ether (250 mL).
  • Alternate Recrystallization Procedure for XIV-II
  • Methyl tertiary-butyl ether (400 mL per 20 g of starting material XIII) was added to the crude product and the pH of the resulting mixture was adjusted to 8-9 using CHA at 20° C. Additional 200 mL methyl tertiary-butyl ether was added and the mixture was mixed at this temperature for 2 h after which it was filtered and the resulting solid was washed with methyl tertiary-butyl ether (10 mL).
  • Alternate Recrystallization Procedure for XIV-III
  • Methyl tertiary-butyl ether (50 mL per 4 g of starting material XIII) was added to the crude product and the pH of the resulting mixture was adjusted to 8-9 using CHA at 20° C. The mixture was mixed at this temperature for 45 min after which it was filtered and the resulting solid was washed with methyl tertiary-butyl ether (10 mL).
  • Recrystallization Procedure for IIa
  • Chloroform (70 mL) was added to the crude product (25 g) and the resulting mixture was cooled to 0° C. Hexanes (210 mL) were then slowly added so as to maintain the temperature at 0° C. The mixture was further maintained at this temperature for 1 h after which it was filtered cooled and the resulting solid was dried under vacuum at 0° C.
  • Alternate Recrystallization Procedure for IIa-I
  • Chloroform (2200 m L) was added to the crude product (1100 g). Hexanes (6600 L) were then added slowly and the resulting mixture was cooled to less than 0° C. The mixture was further mixed at temperature below 0° C. for 1 h after which it was filtered at less than 0° C. and the resulting solid was dried under vacuum at temperature below 0° C.
  • Example 2 Preparation of crystalline N-Fmoc-(S)-α-methyl-α-aminohept-6-enoic acid Example 2a Preparation of (S)-2-[N—(N′-Boc-prolyl)amino]benzophenone
  • Figure US20140128581A1-20140508-C00029
  • Tetrahydrofuran and 7.5 kg (1.0 equivs.) of Boc-L-proline (Va) were added to a reactor and the resulting solution was cooled to −5° C. 4.2 kg (1.05 equivs.) of N-methylmorpholine were charged, followed by slow addition of 5.3 kg (1.0 equivs.) of isobutyl chloroformate in tetrahydrofuran while maintaining an internal temperature of <5° C. The mixture was allowed to agitate at 20-25° C. for 45-60 minutes and then was analyzed by TLC for completion. A solution of 6.2 kg (0.9 equvs.) of 2-aminobenzophenone/tetrahydrofuran was charged and the mixture was allowed to agitate at 20-25° C. until the reaction was shown to be complete by TLC. The mixture was concentrated to ½ A volume and isopropyl acetate was charged. The organic product layer was then washed with a 5% sodium bicarbonate solution, water was charged, and then pH adjusted to 2.0-2.5 with 25% sulfuric acid. Layers were split and the organic product layer was washed again with water. The organic product solution/layer was then concentrated and crystallized from isopropyl acetate and washed with methyl tert-butyl ether. Product (Vila) was then isolated and dried under heat and vacuum. Yield: 9.3 kg, 75%.
  • Example 2b Preparation of L-Proline-2-Aminobenzophenone amide
  • Figure US20140128581A1-20140508-C00030
  • 9.4 kg (1.0 equivs.) of Boc-L-proline-2-aminobenzophenone amide (Vila) was dissolved into acetonitrile.
  • 1.7 kg (2.0 equivs.) of hydrogen chloride gas were then charged/bubbled into the solution. This mixture was allowed to agitate at 20-25° C. until the reaction was demonstrated to be complete by TLC. Methyl tert-butyl ether was added and a solid product was isolated out of the reaction solution and washed with additional methyl tert-butyl ether. The product (VIIIa) was dried under heat and vacuum. Yield: 7.0 kg, 100%.
  • Example 2c Preparation of (S)-2-[N—(N′-benzylprolyl)amino]benzophenone (L-BPB)
  • Figure US20140128581A1-20140508-C00031
  • 7.1 kg (1.0 equivs.) of L-proline-2-aminobenzophenone amide.HCl (VIIIa) was dissolved into tetrahydrofuran and water. 5.8 kg (2.4 equivs.) of triethylamine were then charged followed by a slow addition of 5.9 kg (1.4 equivs.) of benzyl bromide. The mixture was then allowed to agitate at 20-25° C. until the reaction was complete. Methyl tert-butyl ether and water were added and the solution pH was adjusted to 2.0-2.5 with a 1N hydrochloric acid solution. The mixture was concentrated to remove all the tetrahydrofuran. The product slurry was then isolated and washed with methyl tert-butyl ether. The product (IXa) was dried under heat and vacuum. Yield 7.7 kg, 84.0%.
  • Example 2d Preparation of (S)-Ala-Ni-BPB
  • Figure US20140128581A1-20140508-C00032
  • 7.9 kg (1.0 equivs.) of L-BPB (IXa), 12.1 kg (1.78 equivs.) nickel (II) nitrate hexahydrate, 3.7 kg (2.0 equivs) of L-alanine, and methanol were charged to a reactor. The mixture was heated to 40° C. and a solution of 8.2 kg (8.0 equivs.) of potassium hydroxide/methanol was slowly added while maintaining the internal temperature at <50° C. The reaction mixture was then heated up to 60° C. and allowed to agitate at temperature until the reaction was complete. The mixture was subsequently cooled to 20-25° C. and 8.9 kg (5.0 equivs.) of acetic acid was slowly charged while maintaining the internal temperature at <35° C. The reaction solution was then concentrated to a solid. Tetrahydrofuran and isopropyl acetate were added to dissolve the solids and the organic product layer was washed twice with water. The solution was concentrated again and material crystallized out of tetrahydrofuran and methyl tert-butyl ether. The product was isolated, rinsed with additional methyl tert-butyl ether and analyzed for purity. To improve purity the product (XIa) was recrystallized out of isopropyl alcohol and then isolated, and dried under heat and vacuum. Yield: 6.0 kg, 56.0%.
  • Recrystallization procedure for XIa
  • Methyl tertiary-butyl ether (550 mL per 50 g of starting material L-BPB) was added to the crude product (S)-Ala-Ni-BPB and the slurry was then heated to 50° C. before cooling it to 20° C. The mixture was mixed at 20° C. for 16 h. The mixture was filtered and the resulting solid was washed with methyl tertiary-butyl ether (100 mL) to obtain the crystalline product XIa.
  • Alternate Recrystallization Procedure for XIa-I
  • Methyl tertiary-butyl ether (600 mL per 50 g of starting material L-BPB) was added to the crude product (S)-Ala-Ni-BPB and the slurry was then heated to 50-60° C. and maintained at this temperature for 1 h. The mixture was then filtered at 35° C. and washed with methyl tertiary-butyl ether (100 mL) to obtain the crystalline product XIa.
  • Alternate Recrystallization Procedure for XIa
  • Methyl tertiary-butyl ether (500 mL per 50 g of starting material L-BPB) was added to the crude product (S)-Ala-Ni-BPB and the slurry was then heated to 45-50° C. and maintained at this temperature for 1 h. The mixture was then filtered at 35° C. and washed with methyl tertiary-butyl ether (100 mL) to obtain the crystalline product XIa.
  • Alternate Recrystallization Procedure-III
  • Methyl tertiary-butyl ether (2000 mL per 280 g of starting material L-BPB) was added to the crude product (S)-Ala-Ni-BPB and the slurry was then heated to 45-50° C. and maintained at this temperature for 30 min. The mixture was then cooled to 20° C. and mixed at this temperature for 8 h. The resulting solid was then filtered and washed with methyl tertiary-butyl ether (100 mL).
  • (S)-Ala-Ni-BPB (300 g) was recrystallized by dissolving in THF (450 mL). The mixture was heated to 50° C. for 1 h followed by the addition of methyl tertiary-butyl ether (1500 mL) at 50° C. The resulting mixture was mixed at this temperature for additional 1 h. The slurry was then cooled to 20° C. and mixed at 20° C. for 1 h. The resulting solid was then filtered and washed with methyl tertiary-butyl ether (300 mL) to obtain the crystalline product XIa.
  • Example 2e Preparation of S5-Ni-BPB
  • Figure US20140128581A1-20140508-C00033
  • 5.8 kg (1.0 equivs.) of (S)-Ala-Ni-BPB (Xia) was charged to a reactor and dissolved up into dimethylformamide and cooled to 10° C. 1.2 kg (2.5 equivs.) of sodium hydroxide (powder) was then charged to the same reactor and the mixture was sparged with nitrogen and agitated until a solution formed at 10° C. 3.3 kg (2.0 equivs.) of 5-bromo-1-pentene was then charged to the reactor maintaining the internal temperature of <20° C. The mixture was then allowed to agitate at 20-25° C. until the reaction was complete. Once the reaction was complete, the solution was cooled to 10° C. and 0.4 kg (1.5 equivs.) of acetic acid was charged maintaining an internal temperature of <25° C. Water was then charged, followed by methyl tert-butyl ether, and the organic layer was washed. The organic layer was then washed 2 more times with water and then concentrated. The product (XIIa) was crystallized out of isopropyl acetate, isolated and dried under heat and vacuum. Yield: 2.2 kg, 32.4%.
  • Recrystallization Procedure
  • Isopropyl acetate (200 mL per 12.5 g of starting material XIa) was added to the crude product S5-Ni-BPB and the mixture was mixed at 20° C. for 30 min then hexanes (500 mL) were added. The mixture was further mixed for 30 min following which it was filtered to obtain the crystalline product XIIa.
  • Alternate Recrystallization Procedure-I
  • Isopropyl acetate (80 mL per 39 g of starting material XIa) was added to the crude product S5-Ni-BPB and the mixture was mixed at 20° C. for 2 h. The mixture was filtered and washed with isopropyl acetate (35 mL). The filtrate and the washed were combined and heptanes (170 mL) were added. The resulting slurry was mixed for 1 h, then filtered and washed with heptanes (360 mL) to obtain the crystalline product XIIa.
  • Alternate Recrystallization Procedure-II
  • Isopropyl acetate (1000 mL per 205 g of starting material XIa) was added to the crude product S5-Ni-BPB and the mixture was dissolved at 70-80° C. The solution was cooled to 20° C. and the mixture was mixed at this temperature for 1 h during which no crystallization was observed. The mixture was filtered over celite and the solvent was removed under vacuum at 40° C. Methyl tertiary-butyl ether (1000 mL) was added and the mixture was heated to 60° C. then cooled to 20° C. and mixed for 24 h. The solid was filtered and washed with methyl tertiary-butyl ether (300 mL) and to obtain the crystalline product XIIa.
  • Alternate Recrystallization Procedure-III
  • Ethyl acetate (100 mL per 12.5 g of starting material XIa) was added to the crude product S5-Ni-BPB and the mixture was mixed at 20° C. for 30 min. hexanes (500 mL) were added and the resulting slurry was mixed for further 30 min after which it was filtered to obtain the crystalline product XIIa.
  • Alternate Recrystallization Procedure-IV
  • Methyl tertiary-butyl ether (100 mL per 12.5 g of starting material XIa) was added to the crude product S5-Ni-BPB and the mixture was heated to 45-50° C. Heptanes (400 mL) were added 45-50° C. The resulting slurry was cooled to 20° C. and filtered to obtain the crystalline product XIIa.
  • Example 2f Preparation of (S)-2-Amino-2-methyl-hept-6-enoic acid
  • Figure US20140128581A1-20140508-C00034
  • 2.2 kg (1.0 equivs.) of S5-Ni-BPB (XIIa) was charged to a 15-L chem-glassreactor. Tetrahydrofuran was added and the mixture agitated at 20-25° C. until a solution formed. 1.8 kg (4.5 equvs.) of 32% hydrochloric acid was charged slowly while maintaining an internal temperature of <30° C. The mixture was then allowed to agitate for 6-8 hours at ambient temperature. The mixture was concentrated to remove tetrahydrofuran to yield a slurry. Additional water was added and the slurry was agitated at ambient temperature for 1-2 hours. The solid BPB salts were isolated by filtration and rinsed with additional water followed by methyl tert-butyl ether. The product filtrates were then re-charged to the reactor yielding a tri-phased solution. The lower-most layer was split from the upper two layers. The combined two organic layers were then washed 3× with water and concentrated to an oil. Acetonitrile was added and the mixture was warmed to 70° C. for 30 minutes. The mixture was then cooled to 25-30° C. and the solid product was isolated. The solid filter-cake was washed with acetonitrile and methyl tert-butyl ether, then analyzed for chemical purity. The product was then re-slurried out of additional acetonitrile and washed with acetonitrile and methyl tert-butyl ether. The material (XIIIa) was isolated and dried under heat and vacuum. Yield: 0.585 kg, 80.0%
  • Recrystallization Procedure for XIIIa
  • Acetonitrile (100 mL per 20 g of starting material S5-Ni-BPB (XIIa)) was added to the crude product and the mixture was mixed at 20° C. for 1 h. The mixture was then filtered and washed with acetonitrile (40 mL) to obtain the crystalline product XIIIa.
  • Alternate Recrystallization Procedure for XIIIa-I
  • Acetonitrile (500 mL per 185 g of starting material XIIa) was added to the crude product S5-Ni-BPB and the slurry was dissolved at 45-50° C. The solvent was removed under vacuum at 45-50° C., 500 mL acetonitrile was added and the resulting mixture was heated to 45-50° C. The mixture was then cooled to 35° C., filtered and washed with acetonitrile (50 mL) to obtain the crystalline product XIIIa.
  • Alternate Recrystallization Procedure for XIIIa
  • Acetonitrile (270 mL per 35 g of starting material XIIa) was added and the slurry was heated to 45-50° C.
  • The mixture was then cooled to 20° C. and mixed at this temperature for 2 h. The mixture was then filtered and washed with acetonitrile (50 mL) and methyl tertiary-butyl ether (50 mL) to obtain the crystalline product XIIIa.
  • Alternate Recrystallization Procedure for XIIIa
  • Isopropyl acetate (60 mL per 15 g of XIIIa) was added and the mixture was heated to 70° C. Acetonitrile (180 mL) was added and the resulting mixture was cooled to 20° C. The mixture was filtered and the resulting solid was washed with acetonitrile (50 mL) to obtain the crystalline product XIIIa.
  • Example 2g Preparation of N-Fmoc-(S)-α-methyl-α-aminohept-6-enoic acid
  • Figure US20140128581A1-20140508-C00035
  • 0.585 kg (1.0 equiv.) of 2-amino-2-methyl-hept-6-enoic acid.HCl (XIIIa) was suspended in water and polished filtered to remove trace amounts of L-BPB.HCl from the solution. Methyl tert-butyl ether was added and the aqueous product layer extracted once with methyl tert-butyl ether. The aqueous product layer was re-charged and tetrahydrofuran was added. An aqueous 20% sodium carbonate solution (2.75 equiv.) was charged to the mixture, followed by Fmoc-Onsu (0.95 equiv.). The mixture was allowed to react at 20-25° C., while maintaining the pH between 8.5-9.0 with additional amounts of the 20% sodium carbonate solution until the reaction was complete. The mixture was pH adjusted down to pH 2.0-2.5 with conc. hydrochloric acid. Tetrahydrofuran was distilled off and methyl tert-butyl ether is charged. The layers were separated and the organic layer was washed 3 more times with additional water. The organic layer was then concentrated under vacuum and co-stripped with methyl tert-butyl ether. The organic product layer was concentrated and co-stripped with hexanes to a loose oil. The product (IIIa) was then crystallized out of chloroform and hexanes and dried at <0° C. under a 1.0 cfm nitrogen sweep.
  • Yield: 0.831 kg, 76.0%.
  • Recrystallization Procedure for IIa
  • Chloroform (30 mL per 9 g of starting material XIIIa) was added to the crude product. Hexanes (100 mL) were added and the mixture was cooled to 0° C. The resulting solid was filtered at 0° C. and washed with cold hexanes to obtain the crystalline product IIIa.
  • Recrystallization Procedure for Cyclohexylamine Salt of IIIa
  • Acetonitrile (300 mL per 19.04 g of starting material XIIIa) was added to the crude product and the pH was adjusted to 8-9 using cyclohexylamine at 20° C. The resulting mixture was mixed at 20° C. for 2 h and then filtered and washed with acetonitrile (50 mL) to obtain the crystalline cyclohexylamine salt of
  • Alternate recrystallization procedure for cyclohexylamine salt of IIIa-I
  • Methyl tertiary-butyl ether (200 mL per 5 g of starting material XIIIa) was added to the crude product and the pH was adjusted to 8-9 using cyclohexylamine at 20° C. The resulting mixture was mixed at 20° C. for 1 h and then filtered and washed with methyl tertiary-butyl ether (50 mL) to obtain the crystalline cyclohexylamine salt of IIIa.

Claims (35)

What is claimed is:
1. A crystalline compound of Formula (I) or its crystalline salt:
Figure US20140128581A1-20140508-C00036
wherein:
R1 is C1-C3 alkyl, C1-C3 deuteroalkyl, or C1-C3 haloalkyl;
* is a stereocenter;
n is an integer from 1 to 20;
R2 is —H or a nitrogen protecting group; and
R3 is —H or a protecting or activating group.
2. The crystalline compound of Formula (I) or its crystalline salt of claim 1, wherein R1 is C1-C3 alkyl.
3. The crystalline compound of Formula (I) or its crystalline salt of any preceding claim, wherein R1 is methyl.
4. The crystalline compound of Formula (I) or its crystalline salt of any preceding claim, wherein R2 is selected from the group consisting of:
—H,
9-Fluorenylmethoxycarbonyl (Fmoc),
Trityl (Trt),
4-Methoxytrityl (Mmt),
2-(3,5-Dimethoxyphenyl)propan-2-yloxycarbonyl (Ddz),
2-(p-Biphenylyl)-2-propyloxycarbonyl (Bpoc),
2-(4-Nitrophenylsulfonyl)ethoxycarbonyl (NSC),
(1,1-Dioxobenzo[b]thiophene-2-yl)methyloxycarbonyl (Bsmoc),
(1,1-Dioxonaphtho[1,2-b]thiophene-2-yl)methyloxycarbonyl (α-Nsmoc),
1-(4,4-dimethyl-2,6-dioxocyclohex-1-ylidene)-3-methylbutyl (ivDde),
2,-Di-tert-butyl-Fmoc (Fmoc*),
2-Fluoro-Fmoc (Fmoc(2F)),
2-Monoisooctyl-Fmoc (mio-Fmoc),
2,7-Diisooctyl-Fmoc (dio-Fmoc),
2-[Phenyl(methyl)sulfonio]ethyloxy carbonyl tetrafluoroborate (Pms),
Ethanesulfonylethoxycarbonyl (Esc),
2-(4-Sulfophynylsulfonyl)ethoxy carbonyl (Sps),
Tert-butyloxycarbonyl (Boc),
Benzyloxycarbonyl (Z),
Allyloxycarbonyl (Alloc),
2,2,2-Trichloroethyloxycarbonyl (Troc),
p-Nitrobenzyloxycarbonyl (pNZ),
Propargyloxycarbonyl (Poc),
o-Nitrobenzenesulfonyl (oNBS),
2,4-Dinitrobenzenesulfonyl (dNBS),
Benzothiazole-2-sulfonyl (Bts),
o-Nitrobenzyloxycarbonyl (oNz),
4-Nitroveratryloxycarbonyl (NVCO),
2-(2-Nitrophenyl)propyloxycarbonyl (NPPOC),
2,(3,4-Methylethenedioxy-6-nitrophenyl)propyloxycarbonyl (MNPPOC),
9-(4-Bromophenyl)-9-fluorenyl (BrPhF),
Azidomethoxycarbonyl (Azoc),
Hexafluoroacetone (HFA),
2-Chlorobenzyloxycarbonyl (Cl-Z),
4-Methyltrityl (Mtt),
Trifluoroacetyl (tfa),
(Methylsulfonyl)ethoxycarbonyl (Msc),
Phenyldisulphanylethyloxycarbonyl (Phdec),
2-Pyridyldisulphanylethyloxycarbonyl (Pydec), and
o-Nitrobenzenesulfonyl (O-NBS).
5. The crystalline compound of Formula (I) or its crystalline salt of any preceding claim, wherein R2 is selected from the group consisting of:
9-Fluorenylmethoxycarbonyl (Fmoc),
Trityl (Trt),
4-Methoxytrityl (Mmt),
2-(3,5-dimethoxyphenyl)propan-2-yloxycarbonyl (Ddz),
2-(p-biphenylyl)-2-propyloxycarbonyl (Bpoc),
2-(4-Nitrophenylsulfonyl)ethoxycarbonyl (NSC),
1,1-Dioxobenzo[b]thiophene-2-yl)methyloxycarbonyl (Bsmoc),
1-(4,4-dimethyl-2,6-dioxocyclohex-1-ylidene)-3-methylbutyl (ivDde),
Tert-butyloxycarbonyl (Boc),
Benzyloxycarbonyl (Z),
Allyloxycarbonyl (Alloc),
2,2,2-Trichloroethyloxycarbonyl (Troc),
p-Nitrobenzyloxycarbonyl (pNZ),
o-Nitrobenzenesulfonyl (oNBS),
2,4-Dinitrobenzenesulfonyl (dNBS),
o-Nitrobenzyloxycarbonyl (oNz),
4-Nitroveratryloxycarbonyl (NVCO),
2-(2-Nitrophenyl)propyloxycarbonyl (NPPOC),
Hexafluoroacetone (HFA),
2-Chlorobenzyloxycarbonyl (Cl-Z),
4-Methyltrityl (Mtt),
Trifluoroacetyl (tfa),
(Methylsulfonyl)ethoxycarbonyl (Msc), and
o-Nitrobenzenesulfonyl (O-NBS).
6. The crystalline compound of Formula (I) or its crystalline salt of any preceding claim, wherein R2 is selected from the group consisting of:
9-Fluorenylmethoxycarbonyl (Fmoc),
Trityl (Trt),
4-Methoxytrityl (Mmt),
2-(3,5-dimethoxyphenyl)propan-2-yloxycarbonyl (Ddz),
2-(p-biphenylyl)-2-propyloxycarbonyl (Bpoc),
Tert-butyloxycarbonyl (Boc),
Benzyloxycarbonyl (Z),
Allyloxycarbonyl (Alloc),
2,2,2-Trichloroethyloxycarbonyl (Troc),
o-Nitrobenzenesulfonyl (oNBS),
Trityl (Trt),
4-Methyltrityl (Mtt), and
o-Nitrobenzenesulfonyl (O-NBS).
7. The crystalline compound of Formula (I) or its crystalline salt of any preceding claim, wherein R2 is:
9-Fluorenylmethoxycarbonyl (Fmoc).
8. The crystalline compound of Formula (I) or its crystalline salt of any preceding claim, wherein R3 is selected from the group consisting of:
—H,
tert-Butyl (tBu),
2-Chlorotrityl (2-Cl-Trt),
2,4-Dimethoxybenzyl (DMB),
Benzyl (Bn),
2-Phenylisopropyl (2-PhiPr),
5-Phenyl-3,4-ethylenedioxythenyl,
9-Fluorenylmethyl (Fm),
4-(N-[1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl]-amino)benzyl (Dmab),
Methyl (Me),
Ethyl (Et),
Carbamoylmethyl (Cam),
Allyl (Al),
Phenacyl (Pac),
p-Nitrobenzyl (pNB),
2-Trimethylsilylethyl (TMSE),
(2-Phenyl-2-trimethylsilyl)ethyl (PTMSE),
2-(Trimethylsilyl)isopropyl (Tmsi),
Trimethylsilyl (TMS),
2,2,2-Trichloroethyl (Tce),
p-Hydroxyphenacyl (pHP),
4,5-Dimethoxy-2-nitrobenzyl (Dmnb),
1,1-Dimethylallyl (Dma),
Pentaamine cobalt (III),
Succinimide,
p-Nitrophenyl,
Pentafluorophenyl, and
2,4,5-trichlorophenyl.
9. The crystalline compound of Formula (I) or its crystalline salt of any preceding claim, wherein R3 is:
—H.
10. The crystalline compound of Formula (I) or its crystalline salt of any preceding claim, wherein n is an integer from 3 to 11.
11. The crystalline compound of Formula (I) or its crystalline salt of any preceding claim, wherein n is selected from the group consisting of: 3 and 6.
12. The crystalline compound of Formula (I) or its crystalline salt of any of claims 1-11, wherein the stereocenter * is (S).
13. The crystalline compound of Formula (I) or its crystalline salt of any of claims 1-11, wherein the stereocenter * is (R).
14. The crystalline compound of Formula (I) or its crystalline salt of any of claims 1-13, having a chemical purity ranging from about 90% to 100%, optionally determined by HPLC.
15. The crystalline compound of Formula (I) or its crystalline salt of any of claims 1-13, having an optical purity ranging from about 90% to 100%.
16. The crystalline compound of Formula (I) or its crystalline salt of any of claims 1-13, having an optical purity ranging from about 95% to 100%.
17. The crystalline compound of Formula (I) or its crystalline salt of any of claims 1-13, having an enantiomeric excess ranging from about 90% to 100%.
18. The crystalline compound of Formula (I) or its crystalline salt of any of claims 1-13, having an enantiomeric excess ranging from about 95% to 100%.
19. The crystalline compound of Formula (I) or its crystalline salt of claim 1, having the Formula (IIa):
Figure US20140128581A1-20140508-C00037
which optionally has an enantiomeric excess of about 95% to 100%.
20. The crystalline compound of Formula (I) or its crystalline salt of claim 1, having the Formula (IIb):
Figure US20140128581A1-20140508-C00038
which optionally has an enantiomeric excess of about 95% to 100%.
21. The crystalline compound of Formula (I) or its crystalline salt of claim 1, having the Formula (IIIa):
Figure US20140128581A1-20140508-C00039
which optionally has an enantiomeric excess of about 95% to 100%.
22. The crystalline compound of Formula (I) or its crystalline salt of claim 1, having the Formula (IIIb):
Figure US20140128581A1-20140508-C00040
which optionally has an enantiomeric excess of about 95% to 100%.
23. A method of preparing the crystalline compound of Formula (I) or its crystalline salt of claim 1, comprising performing at least one purification step selected from the group consisting of the steps:
1) Crystallizing a metal complex of Formula (XIb)
Figure US20140128581A1-20140508-C00041
from one or more solvents, wherein R1 is C1-C3 alkyl, C1-C3 deuteroalkyl, or C1-C3 haloalkyl, * and ** are each independently stereocenters, and R′, R″, R′″, R″″, and R′″″ are, in the order going around the aromatic ring from R′ to R′″″, selected from the following combinations:
H, H, Cl, Cl, H;
F, F, F, F, F;
F, F, OiPr, F, F;
F, F, OMe, F, F;
Cl, H, H, H, H; or
H, H, Me, Me, H;
2) Precipitating a compound of Formula (Ia) as its HCl salt:
Figure US20140128581A1-20140508-C00042
wherein R1 is C1-C3 alkyl, C1-C3 deuteroalkyl, or C1-C3 haloalkyl, n is an integer from 1 to 20, and * is a stereocenter;
3) Forming an addition salt of Formula (XIVb):
Figure US20140128581A1-20140508-C00043
wherein R1 is C1-C3 alkyl, C1-C3 deuteroalkyl, or C1-C3 haloalkyl, R2 is a nitrogen protecting group, n is an integer from 1 to 20, and * is a stereocenter; and/or
4) Crystallizing a compound of Formula (I) or a salt thereof from one or more solvents, optionally chloroform and/or hexanes.
24. The method of claim 23, comprising performing steps 2), 3), and 4).
25. The method of claim 23, comprising steps 2) and 4).
26. A method of making a polypeptide, comprising making the polypeptide with one or more crystalline compounds of Formula (I) or their crystalline salts of claim 1.
27. The method of claim 23, wherein crystallization of the compound of Formula XIb in step 1 is conducted with methyl tertiary-butyl ether.
28. The method of claim 23, wherein crystallization of the compound Formula XIb in step 1 is conducted with tetrahydrofuran and methyl tertiary-butyl ether.
29. The method of claim 23, wherein crystallization of the compound of Formula XIb in step 1 is conducted with isopropyl acetate.
30. The method of claim 23, wherein the precipitated HCl salt of Formula (Ia) in step 2 is further crystallized with acetonitrile.
31. The method of claim 23, wherein the precipitated HCl salt of Formula (Ia) in step 2 is further crystallized with acetonitrile and methyl tertiary-butyl ether.
32. The method of claim 23, wherein the precipitated HCl salt of Formula (Ia) in step 2 is further crystallized with isopropyl acetate.
33. The method of claim 23, wherein step 3 is conducted in methyl tertiary-butyl ether.
34. The method of claim 23, wherein step 3 is conducted in acetonitrile.
35. The method of claim 23, wherein step 4 comprises crystallizing a CHA salt of Formula (I) with tetrahydrofuran and methyl tertiary-butyl ether.
US14/070,306 2012-11-01 2013-11-01 Disubstituted amino acids and methods of preparation and use thereof Active US9604919B2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/070,306 US9604919B2 (en) 2012-11-01 2013-11-01 Disubstituted amino acids and methods of preparation and use thereof
US15/278,824 US9845287B2 (en) 2012-11-01 2016-09-28 Disubstituted amino acids and methods of preparation and use thereof
US15/794,355 US10669230B2 (en) 2012-11-01 2017-10-26 Disubstituted amino acids and methods of preparation and use thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201261721457P 2012-11-01 2012-11-01
US201361799917P 2013-03-15 2013-03-15
US14/070,306 US9604919B2 (en) 2012-11-01 2013-11-01 Disubstituted amino acids and methods of preparation and use thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/278,824 Continuation US9845287B2 (en) 2012-11-01 2016-09-28 Disubstituted amino acids and methods of preparation and use thereof

Publications (2)

Publication Number Publication Date
US20140128581A1 true US20140128581A1 (en) 2014-05-08
US9604919B2 US9604919B2 (en) 2017-03-28

Family

ID=50622936

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/070,306 Active US9604919B2 (en) 2012-11-01 2013-11-01 Disubstituted amino acids and methods of preparation and use thereof
US15/278,824 Active US9845287B2 (en) 2012-11-01 2016-09-28 Disubstituted amino acids and methods of preparation and use thereof
US15/794,355 Active US10669230B2 (en) 2012-11-01 2017-10-26 Disubstituted amino acids and methods of preparation and use thereof

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/278,824 Active US9845287B2 (en) 2012-11-01 2016-09-28 Disubstituted amino acids and methods of preparation and use thereof
US15/794,355 Active US10669230B2 (en) 2012-11-01 2017-10-26 Disubstituted amino acids and methods of preparation and use thereof

Country Status (13)

Country Link
US (3) US9604919B2 (en)
EP (1) EP2914256B1 (en)
JP (2) JP6526563B2 (en)
KR (1) KR20150082307A (en)
CN (1) CN104812384B (en)
AU (1) AU2013337388B2 (en)
BR (1) BR112015009470A2 (en)
CA (1) CA2887285A1 (en)
IL (1) IL238084A0 (en)
MX (1) MX2015005244A (en)
SG (1) SG11201503052RA (en)
WO (1) WO2014071241A1 (en)
ZA (1) ZA201502991B (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9096684B2 (en) 2011-10-18 2015-08-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9175056B2 (en) 2006-12-14 2015-11-03 Alleron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
US9175045B2 (en) 2008-09-22 2015-11-03 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9458202B2 (en) 2008-11-24 2016-10-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles with improved properties
US9493509B2 (en) 2007-02-23 2016-11-15 Aileron Therapeutics, Inc. Triazole macrocycle systems
US9505804B2 (en) 2012-02-15 2016-11-29 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9845287B2 (en) 2012-11-01 2017-12-19 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US9957299B2 (en) 2010-08-13 2018-05-01 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10023613B2 (en) 2015-09-10 2018-07-17 Aileron Therapeutics, Inc. Peptidomimetic macrocycles as modulators of MCL-1
US10022422B2 (en) 2009-01-14 2018-07-17 Alleron Therapeutics, Inc. Peptidomimetic macrocycles
US10039809B2 (en) 2013-12-18 2018-08-07 The California Institute For Biomedical Research Modified therapeutic agents, stapled peptide lipid conjugates, and compositions thereof
US10059741B2 (en) 2015-07-01 2018-08-28 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10227380B2 (en) 2012-02-15 2019-03-12 Aileron Therapeutics, Inc. Triazole-crosslinked and thioether-crosslinked peptidomimetic macrocycles
US10253067B2 (en) 2015-03-20 2019-04-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10300109B2 (en) 2009-09-22 2019-05-28 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10301351B2 (en) 2007-03-28 2019-05-28 President And Fellows Of Harvard College Stitched polypeptides
US10471120B2 (en) 2014-09-24 2019-11-12 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10905739B2 (en) 2014-09-24 2021-02-02 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and formulations thereof
US10987427B2 (en) 2013-09-13 2021-04-27 The Scripps Research Institute Modified therapeutic agents and compositions thereof

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2558928T3 (en) 2007-01-31 2016-02-09 Dana-Farber Cancer Institute, Inc. Stabilized p53 peptides and uses thereof
WO2014138429A2 (en) 2013-03-06 2014-09-12 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and use thereof in regulating hif1alpha
CN110869044A (en) 2017-02-22 2020-03-06 倍奥英赛普特有限责任公司 Peptides and methods of treating malnutrition-related disorders with peptides
EP4082557A3 (en) 2017-07-17 2023-01-25 Bioincept LLC Peptides and methods of transplantation and restorative organ function
CN107652216A (en) * 2017-11-10 2018-02-02 南京红杉生物科技有限公司 A kind of synthetic method of R 2 [N (N benzyl dried meat acid amides) amino] benzophenone
WO2020023502A1 (en) 2018-07-23 2020-01-30 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7786072B2 (en) * 1999-05-18 2010-08-31 President And Fellows Of Harvard College Stabilized compounds having secondary structure motifs

Family Cites Families (578)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4000259A (en) 1975-06-16 1976-12-28 American Home Products Corporation Cyclic dodecapeptide analogs of somatostatin and intermediates
US4438270A (en) 1977-07-11 1984-03-20 Merrell Toraude Et Compagnie α-Halomethyl derivatives of α-amino acids
US4191754A (en) 1979-02-28 1980-03-04 Merck & Co., Inc. Bicyclic somatostatin analogs
US4270537A (en) 1979-11-19 1981-06-02 Romaine Richard A Automatic hypodermic syringe
AU550730B2 (en) 1982-03-09 1986-04-10 Commonwealth Of Australia, The Automated metal detection
US4728726A (en) 1982-10-04 1988-03-01 The Salk Institute For Biological Studies GRF analogs IIIb
US4518586A (en) 1983-01-13 1985-05-21 The Salk Institute For Biological Studies GRF Analogs III
US5416073A (en) 1983-08-10 1995-05-16 The Adminstrators Of The Tulane Educational Fund Growth hormone-releasing peptides and method of treating animals, therewith
US4596556A (en) 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US5036045A (en) 1985-09-12 1991-07-30 The University Of Virginia Alumni Patents Foundation Method for increasing growth hormone secretion
US4730006A (en) 1986-01-27 1988-03-08 Merrell Dow Pharmaceuticals Inc. Derivatives of 2,6-diamino-3-haloheptanedioic acid
US4880778A (en) 1986-05-12 1989-11-14 Eastman Kodak Company Combinations having synergistic growth hormone releasing activity and methods for use thereof
NL194729C (en) 1986-10-13 2003-01-07 Novartis Ag Process for the preparation of peptide alcohols via solid phase.
CA1283827C (en) 1986-12-18 1991-05-07 Giorgio Cirelli Appliance for injection of liquid formulations
GB8704027D0 (en) 1987-02-20 1987-03-25 Owen Mumford Ltd Syringe needle combination
US5112808A (en) 1987-05-11 1992-05-12 American Cyanamid Company Alkylated hormone-releasing peptides and method of treatig mammals therewith
US4790824A (en) 1987-06-19 1988-12-13 Bioject, Inc. Non-invasive hypodermic injection device
US4940460A (en) 1987-06-19 1990-07-10 Bioject, Inc. Patient-fillable and non-invasive hypodermic injection device assembly
US4941880A (en) 1987-06-19 1990-07-17 Bioject, Inc. Pre-filled ampule and non-invasive hypodermic injection device assembly
US5877277A (en) 1987-09-24 1999-03-02 Biomeasure, Inc. Octapeptide bombesin analogs
US5453418A (en) 1988-03-07 1995-09-26 Eli Lilly And Company Ractopamine and growth hormone combinations
US5339163A (en) 1988-03-16 1994-08-16 Canon Kabushiki Kaisha Automatic exposure control device using plural image plane detection areas
AU3439589A (en) 1988-03-24 1989-10-16 Terrapin Diagnostics, Inc. Molecular sticks for controlling protein conformation
US5094951A (en) 1988-06-21 1992-03-10 Chiron Corporation Production of glucose oxidase in recombinant systems
US5043322A (en) 1988-07-22 1991-08-27 The Salk Institute For Biological Studies Cyclic GRF analogs
FR2638359A1 (en) 1988-11-03 1990-05-04 Tino Dalto SYRINGE GUIDE WITH ADJUSTMENT OF DEPTH DEPTH OF NEEDLE IN SKIN
US5384309A (en) 1989-07-17 1995-01-24 Genentech, Inc. Cyclized peptides and their use as platelet aggregation inhibitors
US5120859A (en) 1989-09-22 1992-06-09 Genentech, Inc. Chimeric amino acid analogues
US5712418A (en) 1989-10-23 1998-01-27 Research Corporation Technologies, Inc. Synthesis and use of amino acid fluorides as peptide coupling reagents
US5650133A (en) 1990-01-19 1997-07-22 Nycomed Salutar Macrocyclic polyaza dichelates linked through ring nitrogens via an amide or ester functionality
US5296468A (en) 1989-10-30 1994-03-22 The Salk Institute For Biological Studies GnRH analogs
US5580957A (en) 1989-10-30 1996-12-03 The Salk Institute For Biological Studies GnRH analogs
US5169932A (en) 1989-10-30 1992-12-08 The Salk Institute For Biological Studies Gnrh analogs
US5352796A (en) 1989-10-30 1994-10-04 The Salk Institute For Biological Studies Amino acids useful in making GnRH analogs
US5064413A (en) 1989-11-09 1991-11-12 Bioject, Inc. Needleless hypodermic injection device
US5312335A (en) 1989-11-09 1994-05-17 Bioject Inc. Needleless hypodermic injection device
US5245009A (en) 1990-03-23 1993-09-14 The Salk Institute For Biological Studies CRF antagonists
CA2047042A1 (en) 1990-07-19 1992-01-20 John Hannah Cyclic hiv principal neutralizing determinant peptides
US5629020A (en) 1994-04-22 1997-05-13 Emisphere Technologies, Inc. Modified amino acids for drug delivery
US6331318B1 (en) 1994-09-30 2001-12-18 Emisphere Technologies Inc. Carbon-substituted diketopiperazine delivery systems
US5714167A (en) 1992-06-15 1998-02-03 Emisphere Technologies, Inc. Active agent transport systems
US5190521A (en) 1990-08-22 1993-03-02 Tecnol Medical Products, Inc. Apparatus and method for raising a skin wheal and anesthetizing skin
SK32693A3 (en) 1990-10-11 1993-09-09 Boehringer Ingelheim Kg Cyclopeptides, a method of preparing them and their use as drugs
DE69118826T2 (en) 1990-11-27 1996-11-14 Fuji Photo Film Co Ltd Propenamide derivatives, their polymers, copolymers and their use
US5124454A (en) 1990-11-30 1992-06-23 Minnesota Mining And Manufacturing Company Polycyclic diamines and method of preparation
US5527288A (en) 1990-12-13 1996-06-18 Elan Medical Technologies Limited Intradermal drug delivery device and method for intradermal delivery of drugs
CA2103577A1 (en) 1991-02-07 1992-08-08 Michael Kahn Conformationally restricted mimetics of beta turns and beta bulges and peptides containing the same
US5747469A (en) 1991-03-06 1998-05-05 Board Of Regents, The University Of Texas System Methods and compositions comprising DNA damaging agents and p53
US5744450A (en) 1991-03-14 1998-04-28 The Salk Institute For Biological Studies GnRH analogs
JPH05507939A (en) 1991-04-09 1993-11-11 エフ・ホフマン―ラ ロシユ アーゲー Analogs of growth hormone releasing factor
US5262519A (en) 1991-05-15 1993-11-16 The Salk Institute For Biological Studies GRF analogs XI
US5364851A (en) 1991-06-14 1994-11-15 International Synthecon, Llc Conformationally restricted biologically active peptides, methods for their production and uses thereof
CA2072249C (en) 1991-06-28 2003-06-17 Saiko Hosokawa Human monoclonal antibody specifically binding to surface antigen of cancer cell membrane
GB9114949D0 (en) 1991-07-11 1991-08-28 Smithkline Beecham Plc Novel compounds
ATE155486T1 (en) 1991-08-13 1997-08-15 Takeda Chemical Industries Ltd CYCLIC PEPTIDES AND THEIR USE
US7517644B1 (en) 1991-08-23 2009-04-14 Larry J. Smith Method and compositions for cellular reprogramming
GB9118204D0 (en) 1991-08-23 1991-10-09 Weston Terence E Needle-less injector
SE9102652D0 (en) 1991-09-13 1991-09-13 Kabi Pharmacia Ab INJECTION NEEDLE ARRANGEMENT
IL103252A (en) 1991-09-30 1997-03-18 Du Pont Merck Pharma CYCLIC COMPOUNDS USEFUL AS INHIBITORS OF PLATELET GLYCOPROTEIN IIb/IIIa AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
ES2133295T3 (en) 1991-11-19 1999-09-16 Takeda Chemical Industries Ltd CYCLIC PEPTIDES AND THEIR USE.
US5328483A (en) 1992-02-27 1994-07-12 Jacoby Richard M Intradermal injection device with medication and needle guard
JP3067031B2 (en) 1992-04-03 2000-07-17 カリフォルニア インスティチュート オブ テクノロジー Olefin metathesis polymerization method
US5411860A (en) 1992-04-07 1995-05-02 The Johns Hopkins University Amplification of human MDM2 gene in human tumors
EP0643726B1 (en) 1992-05-26 1999-08-18 Rijksuniversiteit te Leiden PEPTIDES OF HUMAN p53 PROTEIN FOR USE IN HUMAN T CELL RESPONSE INDUCING COMPOSITIONS, AND HUMAN p53 PROTEIN-SPECIFIC CYTOTOXIC T-LYMPHOCYTES
US5323907A (en) 1992-06-23 1994-06-28 Multi-Comp, Inc. Child resistant package assembly for dispensing pharmaceutical medications
DE69332377T2 (en) 1992-07-13 2003-07-03 Bionebraska Inc METHOD FOR MODIFYING RECOMBINANT POLYPEPTIDES
US5383851A (en) 1992-07-24 1995-01-24 Bioject Inc. Needleless hypodermic injection device
US5569189A (en) 1992-09-28 1996-10-29 Equidyne Systems, Inc. hypodermic jet injector
US5334144A (en) 1992-10-30 1994-08-02 Becton, Dickinson And Company Single use disposable needleless injector
US5371070A (en) 1992-11-09 1994-12-06 The Salk Institute For Biological Studies Bicyclic GnRH antagonists and a method for regulating the secretion of gonadotropins
MX9401351A (en) 1993-02-22 1994-08-31 Alza Corp COMPOSITIONS FOR ORAL SUPPLY FOR ACTIVE AGENTS.
AU6415894A (en) 1993-03-29 1994-10-24 Du Pont Merck Pharmaceutical Company, The Cyclic compounds useful as inhibitors of platelet glycoprotein iib/iiia
US5643957A (en) 1993-04-22 1997-07-01 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
CA2161108A1 (en) 1993-04-23 1994-11-10 Herbert J. Evans Polypeptides that include conformation-constraining groups which flank a protein-protein interaction site
US5446128A (en) 1993-06-18 1995-08-29 The Board Of Trustees Of The University Of Illinois Alpha-helix mimetics and methods relating thereto
ES2229045T3 (en) 1993-08-09 2005-04-16 Sod Conseils Rech Applic DERIVATIVES OF THERAPEUTIC PEPTIDES.
US5622852A (en) 1994-10-31 1997-04-22 Washington University Bcl-x/Bcl-2 associated cell death regulator
US5536814A (en) 1993-09-27 1996-07-16 La Jolla Cancer Research Foundation Integrin-binding peptides
EP0734439A4 (en) 1993-11-22 2000-07-12 Onyx Pharma Inc p53-BINDING POLYPEPTIDES AND POLYNUCLEOTIDES ENCODING SAME
US6287787B1 (en) 1993-11-24 2001-09-11 Torrey Pines Institute For Molecular Studies Dimeric oligopeptide mixture sets
WO1995022546A1 (en) 1994-02-18 1995-08-24 Cell Therapeutics, Inc. Intracellular signalling mediators
WO1995024176A1 (en) 1994-03-07 1995-09-14 Bioject, Inc. Ampule filling device
US5466220A (en) 1994-03-08 1995-11-14 Bioject, Inc. Drug vial mixing and transfer device
US5506207A (en) 1994-03-18 1996-04-09 The Salk Institute For Biological Studies GNRH antagonists XIII
US5824483A (en) 1994-05-18 1998-10-20 Pence Inc. Conformationally-restricted combinatiorial library composition and method
JP3166482B2 (en) 1994-06-07 2001-05-14 日産自動車株式会社 Coloring structure having reflective interference action
IL109943A (en) 1994-06-08 2006-08-01 Develogen Israel Ltd Conformationally constrained backbone cyclized peptide analogs
US6407059B1 (en) 1994-06-08 2002-06-18 Peptor Limited Conformationally constrained backbone cyclized peptide analogs
US5807746A (en) 1994-06-13 1998-09-15 Vanderbilt University Method for importing biologically active molecules into cells
US7553929B2 (en) 1994-06-13 2009-06-30 Vanderbilt University Cell permeable peptides for inhibition of inflammatory reactions and methods of use
US5770377A (en) 1994-07-20 1998-06-23 University Of Dundee Interruption of binding of MDM2 and P53 protein and therapeutic application thereof
US5702908A (en) 1994-07-20 1997-12-30 University Of Dundee Interruption of binding of MDM2 and p53 protein and therapeutic application thereof
AU714966B2 (en) 1994-09-19 2000-01-13 Ricardo J Moro Detection and treatment of cancer
CA2158782C (en) 1994-09-23 2010-01-12 Pierrette Gaudreau Marker for growth hormone-releasing factor receptors
US5681928A (en) 1994-12-16 1997-10-28 The Salk Institute For Biological Studies Betides and methods for screening peptides using same
EP0805819B1 (en) 1994-12-29 2012-02-08 Massachusetts Institute Of Technology Chimeric dna-binding proteins
US5599302A (en) 1995-01-09 1997-02-04 Medi-Ject Corporation Medical injection system and method, gas spring thereof and launching device using gas spring
US5792747A (en) 1995-01-24 1998-08-11 The Administrators Of The Tulane Educational Fund Highly potent agonists of growth hormone releasing hormone
US6169073B1 (en) 1995-02-16 2001-01-02 Bayer Corporation Peptides and peptidomimetics with structural similarity to human p53 that activate p53 function
EP0729972A1 (en) 1995-02-28 1996-09-04 F. Hoffmann-La Roche Ag Peptide derivatives of tetrahydronaphthalene
US5675001A (en) 1995-03-14 1997-10-07 Hoffman/Barrett, L.L.C. Heteroatom-functionalized porphyrazines and multimetallic complexes and polymers derived therefrom
US6514942B1 (en) 1995-03-14 2003-02-04 The Board Of Regents, The University Of Texas System Methods and compositions for stimulating T-lymphocytes
US5700775A (en) 1995-03-24 1997-12-23 Gutniak; Mark K. Method and treatment composition for decreasing patient time in catabolic state after traumatic injury
US5650386A (en) 1995-03-31 1997-07-22 Emisphere Technologies, Inc. Compositions for oral delivery of active agents
CN1151836C (en) 1995-03-31 2004-06-02 艾米斯菲尔技术有限公司 Compound and compositions for delivering active agents
US6090958A (en) 1995-03-31 2000-07-18 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US5731408A (en) 1995-04-10 1998-03-24 Arizona Board Of Regents On Behalf Of The University Of Arizona Peptides having potent antagonist and agonist bioactivities at melanocortin receptors
US6054556A (en) 1995-04-10 2000-04-25 The Arizona Board Of Regents On Behalf Of The University Of Arizona Melanocortin receptor antagonists and agonists
EP0820296A4 (en) 1995-04-14 1999-06-30 Univ Tulane Analogs of growth hormone-releasing factor
US5672584A (en) 1995-04-25 1997-09-30 The University Of Kansas Cyclic prodrugs of peptides and peptide nucleic acids having improved metabolic stability and cell membrane permeability
US6184344B1 (en) 1995-05-04 2001-02-06 The Scripps Research Institute Synthesis of proteins by native chemical ligation
EP0832096B1 (en) 1995-05-04 2001-07-18 The Scripps Research Institute Synthesis of proteins by native chemical ligation
US6020311A (en) 1995-05-26 2000-02-01 Theratechnologies, Inc. GRF analogs with increased biological potency
US5730723A (en) 1995-10-10 1998-03-24 Visionary Medical Products Corporation, Inc. Gas pressured needle-less injection device and method
US6458764B1 (en) 1995-05-26 2002-10-01 Theratechnologies Inc. GRF analogs with increased biological potency
DK0828758T3 (en) 1995-05-26 2002-01-07 Theratechnologies Inc Chimeric adipose pro-GRF analogs with enhanced biological potency
US5817789A (en) 1995-06-06 1998-10-06 Transkaryotic Therapies, Inc. Chimeric proteins for use in transport of a selected substance into cells
US6051554A (en) 1995-06-07 2000-04-18 Peptor Limited Conformationally constrained backbone cyclized somatostatin analogs
US6413994B1 (en) 1999-02-22 2002-07-02 The Salk Institute For Biological Studies Modulators of peroxisome proliferator activated receptor-gamma, and methods for the use thereof
US5811515A (en) 1995-06-12 1998-09-22 California Institute Of Technology Synthesis of conformationally restricted amino acids, peptides, and peptidomimetics by catalytic ring closing metathesis
FR2738151B1 (en) 1995-09-04 1997-09-26 Rhone Poulenc Rorer Sa ANTAGONISTS OF THE ONCOGENIC ACTIVITY OF THE MDM2 PROTEIN, AND THEIR USE IN THE TREATMENT OF CANCERS
EP0766966A3 (en) 1995-09-08 2001-02-28 Eli Lilly And Company Method of treating insulin resistance
US5750499A (en) 1995-10-18 1998-05-12 The Salk Institute For Biological Studies Receptor-selective somatostatin analogs
GB9521544D0 (en) 1995-10-20 1995-12-20 Univ Dundee Activation of P53 protein and therapeutic applications thereof
US5840833A (en) 1995-10-27 1998-11-24 Molecumetics, Ltd Alpha-helix mimetics and methods relating thereto
US6123964A (en) 1995-10-27 2000-09-26 Merck & Co., Inc. Wet granulation formulation of a growth hormone secretagogue
CN1211991A (en) 1995-12-22 1999-03-24 诺沃挪第克公司 Compounds with growth hormone releasing properties
US5807983A (en) 1995-12-28 1998-09-15 The Salk Institute For Biological Studies GNRH antagonist betides
US5893397A (en) 1996-01-12 1999-04-13 Bioject Inc. Medication vial/syringe liquid-transfer apparatus
AU6162996A (en) 1996-01-17 1997-08-11 California Institute Of Technology Synthesis of conformationally restricted amino acids, peptides and peptidomimetics by catalytic ring closing metathesis
US5849954A (en) 1996-01-18 1998-12-15 Research Corporation Technologies, Inc. Method of peptide synthesis
US5849691A (en) 1996-02-20 1998-12-15 The United States Of America As Represented By The Department Of Health And Human Services Peptidomimetic inhibitors of cathepsin D and plasmepsins I and II
AU2595697A (en) 1996-03-29 1997-10-22 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
GB9607549D0 (en) 1996-04-11 1996-06-12 Weston Medical Ltd Spring-powered dispensing device
JP3792777B2 (en) * 1996-05-10 2006-07-05 株式会社カネカ Method for producing 1-alkoxycarbonyl-3-phenylpropyl derivative
US6071926A (en) 1996-05-22 2000-06-06 Arch Development Corporation Sleep quality improvement using a growth hormone secretagogue
US5817752A (en) 1996-06-06 1998-10-06 La Jolla Pharmaceutical Company Cyclic polypeptides comprising a thioether linkage and methods for their preparation
US5817627A (en) 1996-06-14 1998-10-06 Theratechnologies Inc. Long-acting galenical formulation for GRF peptides
ES2136458T3 (en) 1996-06-14 1999-11-16 Takeda Chemical Industries Ltd METHOD FOR SEPARATING N-TERMINAL METIONIN.
US5663316A (en) 1996-06-18 1997-09-02 Clontech Laboratories, Inc. BBC6 gene for regulation of cell death
ATE397621T1 (en) 1996-07-05 2008-06-15 Cancer Rec Tech Ltd INHIBITORS OF INTERACTION BETWEEN P53 AND MDM2
US7083983B2 (en) 1996-07-05 2006-08-01 Cancer Research Campaign Technology Limited Inhibitors of the interaction between P53 and MDM2
EP0923539B1 (en) 1996-07-22 2002-06-05 Novo Nordisk A/S Compounds with growth hormone releasing properties
US5955593A (en) 1996-09-09 1999-09-21 Washington University BH3 interacting domain death agonist
US20020064546A1 (en) 1996-09-13 2002-05-30 J. Milton Harris Degradable poly(ethylene glycol) hydrogels with controlled half-life and precursors therefor
US5965703A (en) 1996-09-20 1999-10-12 Idun Pharmaceuticals Human bad polypeptides, encoding nucleic acids and methods of use
GB9619757D0 (en) 1996-09-21 1996-11-06 Knoll Ag Chemical process
US5856445A (en) 1996-10-18 1999-01-05 Washington University Serine substituted mutants of BCL-XL /BCL-2 associated cell death regulator
JP4024309B2 (en) 1996-10-22 2007-12-19 第一三共株式会社 New treatment for infectious diseases
US6271198B1 (en) 1996-11-06 2001-08-07 Genentech, Inc. Constrained helical peptides and methods of making same
WO1998022122A1 (en) 1996-11-21 1998-05-28 Promega Corporation Alkyl peptide amides and applications
US6060513A (en) 1997-02-07 2000-05-09 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
CZ290347B6 (en) 1997-02-07 2002-07-17 Ústav organické chemie a biochemie AV ČR Cyclopeptides stimulating growth hormone release and process of their preparation
US6313088B1 (en) 1997-02-07 2001-11-06 Emisphere Technologies, Inc. 8-[(2-hydroxy-4-methoxy benzoyl) amino]-octanoic acid compositions for delivering active agents
EA199900752A1 (en) 1997-02-20 2000-06-26 Йеда Рисерч Энд Дивелопмент Ко. Лтд. ANTIPATOGENIC SYNTHETIC PEPTIDES AND COMPOSITIONS INCLUDING THEM
US6849428B1 (en) 1997-03-05 2005-02-01 New England Biolabs, Inc. Intein-mediated protein ligation of expressed proteins
US6635740B1 (en) 1997-03-27 2003-10-21 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Ligand/lytic peptide compositions and methods of use
CA2286867A1 (en) 1997-04-11 1998-10-22 Eli Lilly And Company Combinatorial libraries of peptidomimetic macrocycles and processes therefor
GB9708092D0 (en) 1997-04-22 1997-06-11 Univ Dundee Materials and methods relating to inhibiting the interaction of p53 and mdm2
US6329368B1 (en) 1997-05-09 2001-12-11 The Regents Of The University Of California Endocrine modulation with positive modulators of AMPA type glutamate receptors
EP0989136A4 (en) 1997-05-15 2002-10-09 Kyowa Hakko Kogyo Kk Peptides having cyclic structures and exerting p53 protein activty-restoring effect on p53 protein mutants
US5993412A (en) 1997-05-19 1999-11-30 Bioject, Inc. Injection apparatus
US6127341A (en) 1997-06-20 2000-10-03 Novo Nordisk A/S Compounds with growth hormone releasing properties
US6248358B1 (en) 1998-08-25 2001-06-19 Columbia Laboratories, Inc. Bioadhesive progressive hydration tablets and methods of making and using the same
JP2001524301A (en) 1997-09-17 2001-12-04 ザ・ワルター・アンド・エリザ・ホール・インスティテュート・オヴ・メディカル・リサーチ New therapeutic molecules
WO1999016790A1 (en) 1997-09-26 1999-04-08 University Technologies International, Inc. Use of the tumour suppressor gene p33ing1 for modulation of p53 activity and in tumour diagnosis
US6165732A (en) 1997-10-14 2000-12-26 Washington University Method for identifying apoptosis modulating compounds
US6875594B2 (en) 1997-11-13 2005-04-05 The Rockefeller University Methods of ligating expressed proteins
WO1999029343A1 (en) 1997-12-09 1999-06-17 Thomas Jefferson University Method of treating bladder cancer with wild type vaccinia virus
DK1053019T3 (en) 1998-01-07 2004-04-13 Debio Rech Pharma Sa Degradable heterobifunctional polyethylene glycol acrylates and gels and conjugates derived therefrom
IT1298087B1 (en) 1998-01-08 1999-12-20 Fiderm S R L DEVICE FOR CHECKING THE PENETRATION DEPTH OF A NEEDLE, IN PARTICULAR APPLICABLE TO A SYRINGE FOR INJECTIONS
US6030997A (en) 1998-01-21 2000-02-29 Eilat; Eran Acid labile prodrugs
ES2217738T3 (en) 1998-01-29 2004-11-01 Poly-Med Inc. ABSORBABLE MICROPARTICLES.
JP2002501907A (en) 1998-01-29 2002-01-22 キナートン・リミテッド Method for producing absorbable microparticles
JP4394279B2 (en) 1998-03-09 2010-01-06 ジーランド ファーマ アクティーゼルスカブ Pharmacologically active peptide conjugates with reduced propensity to enzymatic hydrolysis
AU767185B2 (en) 1998-03-23 2003-11-06 President And Fellows Of Harvard College Synthesis of compounds and libraries of compounds
HUP0101629A2 (en) 1998-04-15 2003-03-28 Aventis Pharma Inc Process for the preparation of resin-bound cyclic peptides
US6190699B1 (en) 1998-05-08 2001-02-20 Nzl Corporation Method of incorporating proteins or peptides into a matrix and administration thereof through mucosa
WO1999063929A2 (en) 1998-06-08 1999-12-16 Advanced Medicine, Inc. Multibinding inhibitors of microsomal triglyceride transferase protein
US6326354B1 (en) 1998-08-19 2001-12-04 Washington University Modulation of apoptosis with bid
US6194402B1 (en) 1998-09-02 2001-02-27 Merck & Co., Inc. Enhancement of return to independent living status with a growth hormone secretagogue
US7173005B2 (en) 1998-09-02 2007-02-06 Antyra Inc. Insulin and IGF-1 receptor agonists and antagonists
US6572856B1 (en) 1998-09-10 2003-06-03 The University Of Virginia Patent Foundation Methods for the prevention and treatment of cancer using anti-C3b(i) antibodies
US6696063B1 (en) 1998-12-30 2004-02-24 Applied Research Systems Ars Holding N.V. Treatment of HIV-associated dysmorphia/dysmetabolic syndrome (HADDS) with or without lipodystrophy
CA2360671A1 (en) 1999-01-29 2000-08-03 The Board Of Trustees Of The University Of Illinois P53 inhibitors and therapeutic use of the same
US6372490B1 (en) 1999-02-23 2002-04-16 Curagen Corporation Nucleic acid encoding the MDM interacting protein
JP2003503008A (en) 1999-03-01 2003-01-28 バリアジェニックス インコーポレーテッド Methods for targeting RNA molecules
DE60038734T2 (en) 1999-03-29 2009-07-02 The Procter & Gamble Company, Cincinnati Melanocortin receptor Ligands
US6444425B1 (en) 1999-04-02 2002-09-03 Corixa Corporation Compounds for therapy and diagnosis of lung cancer and methods for their use
US6713280B1 (en) 1999-04-07 2004-03-30 Thomas Jefferson University Enhancement of peptide cellular uptake
CA2306720A1 (en) 1999-04-27 2000-10-27 Philip White Supplement for restoring growth hormone levels
EP1623994A3 (en) 1999-05-17 2008-07-16 ConjuChem Biotechnologies Inc. Protection of endogenous therapeutic peptides from peptidase activity through conjugation to blood components
US20090175821A1 (en) 1999-05-17 2009-07-09 Bridon Dominique P Modified therapeutic peptides with extended half-lives in vivo
EP1180016B1 (en) 1999-05-24 2006-09-27 Introgen Therapeutics, Inc. Methods and compositions for non-viral gene therapy for treatment of hyperproliferative diseases
CN1635898A (en) 1999-07-26 2005-07-06 贝勒医学院 Super-active porcine growth hormone releasing hormone analog
US6461634B1 (en) 1999-08-20 2002-10-08 Edward Marshall Food-based delivery of HGH-stimulating and other nutritional supplements
US20080032931A1 (en) 1999-08-25 2008-02-07 Steward Lance E Activatable clostridial toxins
US20020016298A1 (en) 1999-09-01 2002-02-07 Hay Bruce A. Somatostatin antagonists and agonists that act at the sst subtype 2 receptor
US6696418B1 (en) 1999-09-01 2004-02-24 Pfizer Inc. Somatostatin antagonists and agonists that act at the SST subtype 2 receptor
US20030181367A1 (en) 1999-09-27 2003-09-25 O'mahony Daniel Conjugates of membrane translocating agents and pharmaceutically active agents
ATE435871T1 (en) 1999-11-22 2009-07-15 Zymogenetics Inc METHOD FOR FORMING A PEPTIDE RECEPTOR COMPLEX WITH ZSIG33 POLYPEPTIDES.
US6831155B2 (en) 1999-12-08 2004-12-14 President And Fellows Of Harvard College Inhibition of p53 degradation
US6348558B1 (en) 1999-12-10 2002-02-19 Shearwater Corporation Hydrolytically degradable polymers and hydrogels made therefrom
US6579967B1 (en) 1999-12-14 2003-06-17 The Salk Institute For Biological Studies Receptor-selective somatostatin analogs
WO2001044807A1 (en) 1999-12-16 2001-06-21 Chugai Seiyaku Kabushiki Kaisha METHOD OF SCREENING REMEDY FOR CANCER WITH THE USE OF INTERACTION DOMAINS OF p53 AND MORTALIN
GB0018891D0 (en) 2000-08-01 2000-09-20 Novartis Ag Organic compounds
US20010020012A1 (en) 2000-02-01 2001-09-06 Andersen Maibritt Bansholm Use of compounds for the regulation of food intake
DE10009341A1 (en) 2000-02-22 2001-09-06 Florian Kern Method for antigen-specific stimulation of T lymphocytes
US6495674B1 (en) 2000-02-25 2002-12-17 The Salk Institute For Biological Studies Evectins and their use
US20020002198A1 (en) 2000-04-17 2002-01-03 Parr Tyler B. Chemical synergy to elevate growth hormone release in vertebrates
US6495589B2 (en) 2000-04-28 2002-12-17 Pfizer Inc. Somatostatin antagonists and agonists that act at the SST subtype 2 receptor
US6897286B2 (en) 2000-05-11 2005-05-24 Zymogenetics, Inc. Zsig33-like peptides
WO2001087322A2 (en) 2000-05-17 2001-11-22 Bionebraska, Inc. Peptide pharmaceutical formulations
ATE446758T1 (en) 2000-05-31 2009-11-15 Pfizer Prod Inc USE OF GROWTH HORMONE SECRETAGOGENES TO PROMOTE DIGESTIVE MOTILITY
IL143690A0 (en) 2000-06-19 2002-04-21 Pfizer Prod Inc The use of growth hormone secretagogues to treat systemic lupus erythematosus and inflammatory bowel disease
IL143942A0 (en) 2000-06-29 2002-04-21 Pfizer Prod Inc Use of growth hormone secretagogues for treatment of physical performance decline
US7166712B2 (en) 2000-07-12 2007-01-23 Philadelphia, Health And Education Corporation Mammalian MDM2 binding proteins and uses thereof
IL144468A0 (en) 2000-07-27 2002-05-23 Pfizer Prod Inc Use of growth hormone secretagogues for improvement of functional health status
US7049290B2 (en) 2000-07-28 2006-05-23 Universität Zürich Essential downstream component of the wingless signaling pathway and therapeutic and diagnostic applications based thereon
WO2002010195A2 (en) 2000-08-02 2002-02-07 Theratechnologies Inc. Modified peptides with increased potency
US20040228866A1 (en) 2000-08-04 2004-11-18 Ludwig Institute For Cancer Research Suppressor genes
AU2001284942A1 (en) 2000-08-16 2002-02-25 Georgetown University Medical Center Small molecule inhibitors targeted at bcl-2
JP2004507502A (en) 2000-08-30 2004-03-11 ファイザー・プロダクツ・インク Sustained release formulation for growth hormone secretagogue
IL145106A0 (en) 2000-08-30 2002-06-30 Pfizer Prod Inc Intermittent administration of a geowth hormone secretagogue
EP1318827A4 (en) 2000-09-08 2005-09-14 Gryphon Therapeutics Inc Polymer-modified synthetic proteins
US6720330B2 (en) 2000-11-17 2004-04-13 Pfizer Inc. Somatostatin antagonists and agonists that act at the SST subtype 2 receptor
US20030074679A1 (en) 2000-12-12 2003-04-17 Schwartz Robert J. Administration of nucleic acid sequence to female animal to enhance growth in offspring
JP2004530422A (en) 2000-12-19 2004-10-07 ザ ジョンズ ホプキンス ユニバーシティ JFY1 protein induces rapid apoptosis
US20020091090A1 (en) 2000-12-28 2002-07-11 Cole Bridget M. Somatostatin antagonists and agonists
CU23157A1 (en) 2001-01-03 2006-07-18 Ct Ingenieria Genetica Biotech PHARMACEUTICAL COMPOSITION FOR THE TREATMENT OF TISSULAR DANE DUE TO LACK OF BLOOD IRRIGATION ARTERIAL
JP2004520395A (en) 2001-02-02 2004-07-08 コンジュケム,インコーポレーテッド Long-lasting growth hormone releasing factor derivative
CA2439178C (en) 2001-02-23 2013-06-04 Polyphor Ltd. Template-fixed peptidomimetics with antimicrobial activity
GB0104588D0 (en) 2001-02-24 2001-04-11 Univ Dundee Novel p-53 inducible protein
DE10109813A1 (en) 2001-03-01 2002-09-12 Thomas Stanislawski Tumor peptide antigen from human mdm2 proto-oncogene
WO2002072597A2 (en) 2001-03-09 2002-09-19 University Of Louisville Helicomimetics and stabilized lxxll peptidomimetics
US7019109B2 (en) 2001-03-16 2006-03-28 The Salk Institute For Bilogical Studies SSTR1-selective analogs
WO2004037754A2 (en) 2002-10-24 2004-05-06 Dow Global Technologies Inc. Stabilization of olefin metathesis product mixtures
CZ20032707A3 (en) 2001-04-09 2004-01-14 The Administrators Of The Tulane Educational Fund Somatostatin agonists
US6368617B1 (en) 2001-05-15 2002-04-09 Reliv' International, Inc. Dietary supplement
JP4223390B2 (en) 2001-06-05 2009-02-12 エラン・ファルマ・インターナショナル・リミテッド System and method for milling material
WO2003004068A1 (en) 2001-07-06 2003-01-16 Auckland Uniservices Limited Hypertension treatment
EP1422215A4 (en) * 2001-08-08 2005-07-06 Kaneka Corp Process for producing optically active 2-substituted carboxylic acid
AU2002324909A1 (en) 2001-09-07 2003-03-24 Baylor College Of Medicine Linear dna fragments for gene expression
US20040106548A1 (en) 2001-09-07 2004-06-03 Schmidt Michelle A Conformationally constrained labeled peptides for imaging and therapy
US20020045192A1 (en) 2001-09-19 2002-04-18 St. Jude Children's Research Hospital Arf and HDM2 interaction domains and methods of use thereof
EP1312363A1 (en) 2001-09-28 2003-05-21 Pfizer Products Inc. Methods of treatment and kits comprising a growth hormone secretagogue
US20030083241A1 (en) 2001-11-01 2003-05-01 Young Charles W. Use of somatostatin receptor agonists in the treatment of human disorders of sleep hypoxia and oxygen deprivation
CN101157924A (en) 2001-12-11 2008-04-09 人体基因组科学有限公司 Neutrophil leukocyte gene alpha
EP1321474A1 (en) 2001-12-18 2003-06-25 Universite De Geneve A method for inducing apoptosis
WO2003051389A2 (en) 2001-12-18 2003-06-26 Theratechnologies Inc. Pharmaceutical compositions comprising unacylated ghrelin and therapeutical uses thereof
WO2003055510A1 (en) 2001-12-24 2003-07-10 Auckland Uniservices Limited Therapy for growth hormone induced insulin resistance in juveniles with growth disorders
EP1469871A4 (en) 2001-12-31 2006-08-23 Dana Farber Cancer Inst Inc Method of treating apoptosis and compositions thereof
EP1468013A4 (en) 2002-01-03 2005-03-16 Yissum Res Dev Co Conformationally constrained c-backbone cyclic peptides
CA2475273A1 (en) 2002-02-07 2003-08-14 Baylor College Of Medicine Modified pituitary gland development in offspring from expectant mother animals treated with growth hormone releasing hormone therapy
CA2472956A1 (en) 2002-02-14 2003-08-21 Bayer Pharmaceuticals Corporation Formulation strategies in stabilizing peptides in organic solvents and in dried states
WO2003070892A2 (en) 2002-02-15 2003-08-28 The Regents Of The University Of Michigan Inhibitors of rgs proteins
US20030166138A1 (en) 2002-02-21 2003-09-04 Todd Kinsella Cyclic peptides and analogs useful to treat allergies
WO2003081258A2 (en) 2002-03-26 2003-10-02 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with growth hormone secretagogue receptor (ghs)
US7498134B2 (en) 2002-03-30 2009-03-03 The Trustees Of Columbia University In The City Of New York HAUSP-Mdm2 interaction and uses thereof
DE60320780D1 (en) 2002-04-22 2008-06-19 Univ Florida FUNCTIONALIZED NANOPARTICLES AND USE METHOD
WO2003095625A2 (en) 2002-05-13 2003-11-20 3-Dimensional Pharmaceuticals, Inc. Method for cytoprotection through mdm2 and hdm2 inhibition
DE60332725D1 (en) 2002-05-30 2010-07-08 Scripps Research Inst COPPER-CATALYZED LEADING OF AZIDES AND ACETYLENES
WO2003102538A2 (en) 2002-05-30 2003-12-11 European Molecular Biology Laboratory Combinatorial chemical library ii
US7208154B2 (en) 2002-06-03 2007-04-24 Regents Of The University Of Michigan Methods and compositions for the treatment of MHC-associated conditions
SE0201863D0 (en) 2002-06-18 2002-06-18 Cepep Ab Cell penetrating peptides
AU2003253883A1 (en) 2002-07-15 2004-02-02 The Johns Hopkins University Neuronal and optic nerve gene expression patterns
AU2003259172A1 (en) 2002-07-24 2004-02-09 The Salk Institute For Biological Studies Receptor (sstr4)- selective somatostatin analogs
JPWO2004022766A1 (en) 2002-09-06 2005-12-22 株式会社カネカ Method for producing L-α-methylcysteine derivative
AU2003267124A1 (en) 2002-09-09 2004-03-29 Dana-Farber Cancer Institute, Inc. Bh3 peptides and method of use thereof
RU2005111253A (en) 2002-09-18 2005-11-20 Сантр Оспиталье Де Л` Юниверсите Де Монреаль (Схюм) (Ca) GHRH ANALOGUES
AU2003277891A1 (en) 2002-09-23 2004-04-08 Medivir Ab Hcv ns-3 serine protease inhibitors
JP2006517787A (en) 2002-10-07 2006-08-03 ルートヴィヒ・インスティテュート・フォー・カンサー・リサーチ Polypeptide
US20040208866A1 (en) 2002-10-07 2004-10-21 Jaspers Stephen R. Methods of regulating body weight
EP1407779A1 (en) 2002-10-10 2004-04-14 Gastrotech A/S Use of ghrelin for treating low body weight and body fat in gastrectomized individuals
KR100966667B1 (en) 2002-11-07 2010-06-29 코산 바이오사이언시즈, 인코포레이티드 Trans-9,10-dehydroepothilone c and d, analogs thereof and methods of making the same
WO2004041275A1 (en) 2002-11-08 2004-05-21 F. Hoffmann-La Roche Ag Substituted 4-alkoxyoxazol derivatives as ppar agonists
US20050227932A1 (en) 2002-11-13 2005-10-13 Tianbao Lu Combinational therapy involving a small molecule inhibitor of the MDM2: p53 interaction
US7166575B2 (en) 2002-12-17 2007-01-23 Nastech Pharmaceutical Company Inc. Compositions and methods for enhanced mucosal delivery of peptide YY and methods for treating and preventing obesity
EP1578798A1 (en) 2002-12-20 2005-09-28 7TM Pharma A/S Ghrelin receptor inverse agonist for regulation of feeding behaviours
WO2004058804A1 (en) 2002-12-24 2004-07-15 Walter And Eliza Hall Institute Of Medical Research Peptides and therapeutic uses thereof
US20040231909A1 (en) 2003-01-15 2004-11-25 Tai-Yang Luh Motorized vehicle having forward and backward differential structure
US20040204358A1 (en) 2003-01-28 2004-10-14 Advisys, Inc. Reducing culling in herd animals growth hormone releasing hormone (GHRH)
US20050059605A1 (en) 2003-01-31 2005-03-17 Krishna Peri Chemically modified metabolites of regulatory peptides and methods of producing and using same
US7638138B2 (en) 2003-02-21 2009-12-29 Translational Research, Ltd. Compositions for nasal administration of pharmaceuticals
WO2004076483A1 (en) 2003-02-26 2004-09-10 Japan Science And Technology Agency Transcriptional factor inducing apoptosis in cancer cell
EP1452868A2 (en) 2003-02-27 2004-09-01 Pepscan Systems B.V. Method for selecting a candidate drug compound
US20070060512A1 (en) 2003-03-04 2007-03-15 Homayoun Sadeghi Dipeptidyl-peptidase protected protein
WO2004084943A1 (en) 2003-03-19 2004-10-07 Beth Israel Deaconess Medical Center, Inc Use of antagonists of ghrelin or ghrelin receptor to treat intestinal inflammation
US7632920B2 (en) 2003-04-10 2009-12-15 Schering Corporation Soluble, stable form of HDM2, crystalline forms thereof and methods of use thereof
ES2383752T3 (en) 2003-05-15 2012-06-26 Trustees Of Tufts College GLP-1 stable analogs
EP2382984A3 (en) 2003-05-29 2011-11-23 Theratechnologies Inc. GRF analog compositions and their use
AU2003902743A0 (en) 2003-06-02 2003-06-19 Promics Pty Limited Process for the preparation of cyclic peptides
US20050031549A1 (en) 2003-06-09 2005-02-10 Nastech Pharmaceutical Company Inc. Compositions and methods for enhanced mucosal delivery of growth hormone
US20090198050A1 (en) 2003-06-18 2009-08-06 Tranzyme Pharma Inc. Macrocyclic Modulators of the Ghrelin Receptor
JP4928261B2 (en) 2003-06-18 2012-05-09 トランザイム・ファーマ・インコーポレイテッド Macrocyclic antagonist of motilin receptor
US7476653B2 (en) 2003-06-18 2009-01-13 Tranzyme Pharma, Inc. Macrocyclic modulators of the ghrelin receptor
USRE42624E1 (en) 2003-06-18 2011-08-16 Tranzyme Pharma Inc. Methods of using macrocyclic modulators of the ghrelin receptor
US7491695B2 (en) 2003-06-18 2009-02-17 Tranzyme Pharma Inc. Methods of using macrocyclic modulators of the ghrelin receptor
USRE42013E1 (en) 2003-06-18 2010-12-28 Tranzyme Pharma Inc. Macrocyclic modulators of the ghrelin receptor
WO2005000876A2 (en) 2003-06-27 2005-01-06 Proteologics, Inc. Ring finger family proteins and uses related thereto
WO2005007675A2 (en) 2003-07-09 2005-01-27 The Scripps Research Institute TRIAZOLE &epsiv;-AMINO ACIDS
US20070185031A1 (en) 2003-07-14 2007-08-09 Northwestern University Reducing polyglutamine-based aggregation
GB0317815D0 (en) 2003-07-30 2003-09-03 Amersham Health As Imaging agents
CA2536112A1 (en) 2003-08-20 2005-03-03 Northern Sydney And Central Coast Area Health Service Methods for enhancing embryo viability
CN100475271C (en) 2003-08-20 2009-04-08 加利福尼亚大学董事会 Somatostatin analogs with inhibitory activity to growth hormone release
WO2005027913A1 (en) 2003-09-19 2005-03-31 Pfizer Products Inc. Pharmaceutical compositions and methods comprising combinations of 2-alkylidene-19-nor-vitamin d derivatives and a growth hormone secretagogue
ATE396973T1 (en) 2003-10-03 2008-06-15 Merck & Co Inc BENZYL ETHER AND BENZYLAMINO BETA SECRETASE INHIBITORS FOR THE TREATMENT OF ALZHEIMER'S DISEASE
WO2005039546A2 (en) 2003-10-03 2005-05-06 Veijlen N.V. Use of indoleacetic acid derivatives which increase the serum igf-1 level for the preparation of a therapeutical composition for treatment of various diseases
GB0323728D0 (en) 2003-10-10 2003-11-12 Royal College Of Surgeons Ie Peptidomimetics and uses thereof
WO2005040202A2 (en) 2003-10-16 2005-05-06 Aplagen Gmbh Stabilized alpha-helical peptides
WO2005044840A2 (en) 2003-10-17 2005-05-19 The Cbr Institute For Biomedical Research, Inc. Modulation of anergy and methods for isolating anergy-modulating compounds
WO2005037307A1 (en) 2003-10-20 2005-04-28 Theratechnologies Inc. Use of growth hormone releasing factor analogs in treating patients suffering from wasting
US7273927B2 (en) 2003-11-03 2007-09-25 University Of Massachusetts Mdm2 splice variants
ES2586387T3 (en) 2003-11-05 2016-10-14 Dana-Farber Cancer Institute, Inc. Suitable alpha helical peptides to activate or inhibit cell death
US20050147581A1 (en) 2003-11-19 2005-07-07 The Board Of Trustees Of The University Of Illinois Macromolecular drug complexes having improved stability and therapeutic use of the same
WO2005074521A2 (en) 2004-01-30 2005-08-18 The Trustees Of Columbia University In The City Of New York C-TERMINAL p53 PALINDROMIC PEPTIDE THAT INDUCES APOPTOSIS OF CELLS WITH ABERRANT p53 AND USES THEREOF
US20070161551A1 (en) 2004-02-10 2007-07-12 De Luca Giampiero Methods and compositions for the treatment of lipodystrophy
GB0404731D0 (en) 2004-03-03 2004-04-07 Indp Administrative Inst Nims Method and products for the selective degradation of proteins
US20050203009A1 (en) 2004-03-12 2005-09-15 Bayer Pharmaceuticals Corporation VPAC1 selective antagonists and their pharmacological methods of use
EP1737884B1 (en) 2004-03-19 2016-10-19 The University Of Queensland Alpha helical mimics, their uses and methods for their production
KR20070010151A (en) 2004-03-30 2007-01-22 사파이어 세라퓨틱스, 인크. Method of reducing c-reactive protein using growth hormone secretagogues
JP2007532495A (en) 2004-04-07 2007-11-15 ガストロテック・ファルマ・アクティーゼルスカブ Use of secretagogues to treat ghrelin deficiency
US7034050B2 (en) 2004-04-28 2006-04-25 Romano Deghenghi Pseudopeptides growth hormone secretagogues
CA2565189A1 (en) 2004-05-18 2005-11-24 F. Hoffmann-La Roche Ag Cis-2,4,5-triphenyl imidazolines and their use as anti-cancer medicaments
US7202332B2 (en) 2004-05-27 2007-04-10 New York University Methods for preparing internally constrained peptides and peptidomimetics
WO2005118634A2 (en) 2004-06-04 2005-12-15 The Brigham And Women's Hospital, Inc. Helical peptidomimetics with enhanced activity against beta-amyloid production
EP1602663A1 (en) 2004-06-04 2005-12-07 Chiralix B.V. Triazole-linked glycoamino acids and glycopeptides
CN100335467C (en) 2004-06-04 2007-09-05 中国科学院上海有机化学研究所 Synthesis of 5-iodo-1,4-twice substituted-1,2,3-trioxazole compound
US7893278B2 (en) 2004-06-17 2011-02-22 Hoffman-La Roche Inc. CIS-imidazolines
KR101699142B1 (en) 2004-06-18 2017-01-23 암브룩스, 인코포레이티드 Novel antigen-binding polypeptides and their uses
US7674787B2 (en) 2004-07-09 2010-03-09 The Regents Of The University Of Michigan Conformationally constrained Smac mimetics and the uses thereof
PT1789067E (en) 2004-08-12 2012-08-06 Helsinn Healthcare Sa Use of growth hormone secretagogues for stimulating the motility of the gastrointestinal system
US8039456B2 (en) 2004-08-12 2011-10-18 Helsinn Therapeutics (U.S.), Inc. Method of stimulating the motility of the gastrointestinal system using ipamorelin
WO2006023608A2 (en) 2004-08-18 2006-03-02 Elixir Pharmaceuticals, Inc. Growth-hormone secretagogues
US7402652B2 (en) 2004-09-14 2008-07-22 Miller Landon C G Baclofen conjugate and a pharmaceutical composition for treatment of neuronal disorders
US7151084B2 (en) 2004-12-27 2006-12-19 Miller Landon C G Compound and method of treating neurogenic conditions using non-steroidal anti-inflammatory drug complexes
US7074775B2 (en) 2004-09-14 2006-07-11 Miller Landon C G Aminobutyramide conjugate and a pharmaceutical composition for treatment of neuronal disorders
US7157421B2 (en) 2004-12-27 2007-01-02 Miller Landon C G Piracetam and piracetam analog conjugate and a pharmaceutical composition for treatment of neuronal disorders
US9598470B2 (en) 2004-10-07 2017-03-21 Craig W. Beattie Compositions and methods to prevent cancer by stabilizing P53 through non MDM2-mediated pathways
EP1812048A4 (en) 2004-10-20 2012-01-18 Theratechnologies Inc Gh secretagogues and uses thereof
WO2006050034A1 (en) 2004-10-29 2006-05-11 Schering Corporation Substituted 5-carboxyamide pyrazoles and [1,2,4]triazoles as antiviral agents
US7998930B2 (en) 2004-11-04 2011-08-16 Hanall Biopharma Co., Ltd. Modified growth hormones
WO2006069001A2 (en) 2004-12-20 2006-06-29 Baylor College Of Medicine Structural requirements for stat3 binding and recruitment to phosphototyrosine ligands
GB0428187D0 (en) 2004-12-23 2005-01-26 Univ Liverpool Cancer treatment
PT1844337E (en) 2005-01-24 2014-04-03 Pepscan Systems Bv Binding compounds, immunogenic compounds and peptidomimetics
FR2881430B1 (en) 2005-02-01 2010-10-22 Servier Lab NOVEL PEPTIDES INTERACTING WITH ANTI-APOPTOTIC MEMBERS OF THE BCL-2 PROTEIN FAMILY AND USES THEREOF
US7759383B2 (en) 2005-02-22 2010-07-20 The Regents Of The University Of Michigan Small molecule inhibitors of MDM2 and the uses thereof
AU2006225116B2 (en) 2005-03-15 2012-04-19 Allergan, Inc. Modified Clostridial toxins with altered targeting capabilities for Clostridial toxin target cells
WO2006103666A2 (en) 2005-03-28 2006-10-05 Yeda Research And Development Co. Ltd. Isolated bid polypeptides, polynucleotides encoding same and antibodies directed thereagainst and methods of using same for inducing cell cycle arrest or apoptosis
WO2006122931A1 (en) 2005-05-20 2006-11-23 Biovitrum Ab (Publ) Beta-carboline derivatives and theri use as ghsr modulators
JP5122446B2 (en) 2005-06-13 2013-01-16 トランザイム・ファーマ,インコーポレイテッド Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of use thereof
US20090275648A1 (en) 2005-06-13 2009-11-05 Fraser Graeme L Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
US20070020620A1 (en) 2005-07-14 2007-01-25 Finn M G Compositions and methods for coupling a plurality of compounds to a scaffold
WO2007009191A1 (en) 2005-07-21 2007-01-25 Newsouth Innovations Pty Limited Method for treating cancer
US8138218B2 (en) 2005-07-22 2012-03-20 Ipsen Pharma S.A.S. Growth hormone secretagogues
EP1757290A1 (en) 2005-08-16 2007-02-28 Zentaris GmbH Novel triazole derivatives as ghrelin analogue ligands of growth hormone secretagogue receptors
WO2007038678A2 (en) 2005-09-28 2007-04-05 Societe De Conseils De Recherches Et D'applications Scientifiques S.A.S. Analogs of ghrelin
US20070161544A1 (en) 2006-01-06 2007-07-12 Peter Wipf Selective targeting agents for mitcochondria
EP1986673A2 (en) 2006-02-13 2008-11-05 The Government Of The Usa As Represented By The Secretary Of The Dept. Of Health and Human Services Methods and compositions related to ghs-r antagonists
US7538190B2 (en) 2006-02-17 2009-05-26 Polychip Pharmaceuticals Pty Ltd Methods for the synthesis of two or more dicarba bridges in organic compounds
US7745573B2 (en) 2006-02-17 2010-06-29 Polychip Pharmaceuticals Pty Ltd. Conotoxin analogues and methods for synthesis of intramolecular dicarba bridge-containing peptides
GB0603295D0 (en) 2006-02-18 2006-03-29 Ardana Bioscience Ltd Methods and kits
CU23592A1 (en) 2006-02-28 2010-11-11 Ct Ingenieria Genetica Biotech METHOD TO PREVENT AND ELIMINATE FIBROSIS AND OTHER FORMS OF PATHOLOGICAL DEPOSIT IN THE FABRICS APPLYING THE GHRP-6 SECRETAGOGO PEPTIDE
US7981860B2 (en) 2006-03-13 2011-07-19 Liat Mintz Use of ghrelin splice variant for treating cachexia and/or anorexia and/or anorexia-cachexia and/or malnutrition and/or lipodystrophy and/or muscle wasting and/or appetite-stimulation
JP2010523466A (en) 2006-04-13 2010-07-15 グラクソ グループ リミテッド Aryl and heteroaryl sulfonamides as growth hormone secretagogue receptor agonists
WO2007127457A2 (en) 2006-04-28 2007-11-08 The Administrators Of The Tulane Educational Fund Ghrelin/growth hormone releasing peptide/growth hormone secretatogue receptor antagonists and uses thereof
GB0611405D0 (en) 2006-06-09 2006-07-19 Univ Belfast FKBP-L: A novel inhibitor of angiogenesis
US20090326193A1 (en) 2006-06-23 2009-12-31 Aegis Therapeutics Llc Stabilizing alkylglycoside compositions and methods thereof
US8173594B2 (en) 2006-06-23 2012-05-08 Aegis Therapeutics, Llc Stabilizing alkylglycoside compositions and methods thereof
US8084022B2 (en) 2006-06-23 2011-12-27 Aegis Therapeutics, Llc Stabilizing alkylglycoside compositions and methods thereof
US7998927B2 (en) 2006-06-23 2011-08-16 Aegis Therapeutics, Llc Stabilizing alkylglycoside compositions and methods thereof
US8226949B2 (en) 2006-06-23 2012-07-24 Aegis Therapeutics Llc Stabilizing alkylglycoside compositions and methods thereof
US7425542B2 (en) 2006-06-23 2008-09-16 Aegis Therapeutics, Inc. Stabilizing alkylglycoside compositions and methods thereof
EP2037919A2 (en) 2006-06-30 2009-03-25 Schering Corporation Method of using substituted piperidines that increase p53 activity
CA2656398A1 (en) 2006-06-30 2008-01-10 Schering Corporation Substituted piperidines that increase p53 activity and the uses thereof
US8088733B2 (en) 2006-07-06 2012-01-03 Tranzyme Pharma Inc. Methods of using macrocyclic agonists of the ghrelin receptor for treatment of gastrointestinal motility disorders
EP2051725A4 (en) 2006-07-11 2011-06-15 Harkness Pharmaceuticals Inc Methods of treating obesity using satiety factors
AU2007277556B2 (en) 2006-07-26 2012-11-29 Pepscan Systems B.V. Immunogenic compounds and protein mimics
US8614192B2 (en) 2006-07-28 2013-12-24 Leiden University Medical Center Method for treating ocular cancer
US7737174B2 (en) 2006-08-30 2010-06-15 The Regents Of The University Of Michigan Indole inhibitors of MDM2 and the uses thereof
ATE488232T1 (en) 2006-09-04 2010-12-15 Univ Dundee P53 ACTIVATE BENZOYL UREA AND BENZOYLTHIOUREA COMPOUNDS
US20080213175A1 (en) 2006-09-15 2008-09-04 Kolb Hartmuth C Click chemistry-derived cyclic peptidomimetics as integrin markers
WO2008034736A2 (en) 2006-09-21 2008-03-27 F. Hoffmann-La Roche Ag Oxindole derivatives as anticancer agents
US7897394B2 (en) 2006-09-21 2011-03-01 Intrexon Corporation Endoplasmic reticulum localization signals
US8299022B2 (en) 2006-09-27 2012-10-30 Ipsen Pharma S.A.S. Analogs of ghrelin substituted at the N-terminal
EP2517720A1 (en) 2006-10-05 2012-10-31 New York Blood Center, Inc. Stabilized therapeutic small helical antiviral peptides
KR100860060B1 (en) 2006-10-12 2008-09-24 한국과학기술연구원 METHOD FOR QUANTITATIVE ANALYSIS OF INTERACTIONS BETWEEN HIF-1ALPHA C-TERMINAL PEPTIDES AND CBP OR p300 PROTEINS AND METHOD OF SCREENING INHIBITORS AGAINST FORMATION OF PROTEIN COMPLEX USING THE SAME
US8691761B2 (en) 2006-10-16 2014-04-08 Jean E. F. Rivier Somatostatin receptor 2 antagonists
EP2433963B1 (en) 2006-10-16 2014-06-04 The Salk Institute for Biological Studies Receptor (SSTR2)-selective somatostatin antagonists
EP2091552A4 (en) 2006-11-15 2010-01-06 Dana Farber Cancer Inst Inc Stabilized maml peptides and uses thereof
US7932397B2 (en) 2006-11-22 2011-04-26 Massachusetts Institute Of Technology Olefin metathesis catalysts and related methods
US7981998B2 (en) 2006-12-14 2011-07-19 Aileron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
BRPI0720306A2 (en) 2006-12-14 2014-02-04 Aileron Therapeutics Inc BIS-SUFIDRIL MACROCYCLING SYSTEMS
WO2008074895A1 (en) 2006-12-21 2008-06-26 Cytos Biotechnology Ag Circular ccr5 peptide conjugates and uses thereof
EP2114986B1 (en) 2007-01-29 2013-05-22 Polyphor Ltd. Template-fixed peptidomimetics
ES2558928T3 (en) 2007-01-31 2016-02-09 Dana-Farber Cancer Institute, Inc. Stabilized p53 peptides and uses thereof
WO2008130464A1 (en) 2007-02-09 2008-10-30 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor modulators and methods of using the same
CN101663044B (en) 2007-02-23 2014-07-23 爱勒让治疗公司 Triazole macrocycle systems
WO2008106507A2 (en) 2007-02-27 2008-09-04 University Of South Florida Mdm2/mdmx inhibitor peptide
US7816324B2 (en) 2007-03-13 2010-10-19 Board Of Regents, The University Of Texas System Composition and method for the treatment of diseases affected by a peptide receptor
KR101623985B1 (en) * 2007-03-28 2016-05-25 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Stitched polypeptides
TWI429436B (en) 2007-04-10 2014-03-11 Helsinn Therapeutics Us Inc Methods of treating or preventing emesis using growth hormone secretagogues
US20080260820A1 (en) 2007-04-19 2008-10-23 Gilles Borrelly Oral dosage formulations of protease-resistant polypeptides
EP2152294B1 (en) 2007-05-02 2017-07-26 Dana-Farber Cancer Institute, Inc. A bad bh3 domain peptide for use in treating or delaying the onset of diabetes.
AU2008260483A1 (en) 2007-06-01 2008-12-11 Acologix, Inc. High temperature stable peptide formulation
US20090088380A1 (en) 2007-07-12 2009-04-02 Pierrette Gaudreau Ghrh analogs and therapeutic uses thereof
RU2007133287A (en) 2007-09-05 2009-03-10 Ионов Иль Давидович (RU) ANTIPOPSORIATIC AGENT AND METHOD OF ITS USE (OPTIONS)
JP2010538982A (en) 2007-09-11 2010-12-16 モンドバイオテック ラボラトリーズ アクチエンゲゼルシャフト Use of insulin C-peptide as a therapeutic agent, alone or in combination with GLP-1
WO2009033768A2 (en) 2007-09-11 2009-03-19 Mondobiotech Laboratories Ag Use of a peptide as a therapeutic agent
EP2185182A1 (en) 2007-09-11 2010-05-19 Mondobiotech Laboratories AG Use of grf-1 (1-29 ) and corticotropin-releasing factor as therapeutic agents
EP2197431A4 (en) 2007-09-17 2013-03-27 Olas Pharmaceuticals Inc Modulation of growth hormone, dhea, and cortisol with positive modulators of ampa type glutamate receptors
ATE547415T1 (en) 2007-09-21 2012-03-15 Janssen Pharmaceutica Nv INHIBITORS OF THE INTERACTION BETWEEN MDM2 AND P53
JP5783721B2 (en) 2007-09-26 2015-09-24 ダナ ファーバー キャンサー インスティテュート インコーポレイテッド Methods and compositions for modulating BCL-2 family polypeptides
KR101500528B1 (en) 2007-12-03 2015-03-09 이탈파마코 에스.피.에이. New non-selective somatostatin analogues
US8871899B2 (en) 2007-12-31 2014-10-28 New York University Control of viral-host membrane fusion with hydrogen bond surrogate-based artificial helices
EP2235064B1 (en) 2008-01-07 2015-11-25 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
AU2009206684C1 (en) 2008-01-25 2014-12-04 Massachusetts Institute Of Technology Catalysts for metathesis reactons including enantioselective olefin metathesis, and related methods
MX2010008168A (en) 2008-01-25 2011-02-24 P53 Inc P53 biomarkers.
EP2242503A4 (en) 2008-02-08 2012-04-25 Aileron Therapeutics Inc Therapeutic peptidomimetic macrocycles
US20090326192A1 (en) 2008-04-08 2009-12-31 Aileron Therapeutics, Inc. Biologically active peptidomimetic macrocycles
US20110144303A1 (en) 2008-04-08 2011-06-16 Aileron Therapeutics, Inc. Biologically Active Peptidomimetic Macrocycles
WO2009137532A1 (en) 2008-05-06 2009-11-12 New York Blood Center Antiviral cell penetrating peptides
WO2009149214A2 (en) 2008-06-03 2009-12-10 Aileron Therapeutics, Inc. Compositions and methods for enhancing cellular transport of biomolecules
WO2009149339A2 (en) 2008-06-05 2009-12-10 University Of Maryland, Baltimore P53 activator peptides
US8796216B2 (en) 2008-06-12 2014-08-05 Syntaxin Limited Suppression of neuroendocrine diseases
US20110158973A1 (en) 2008-06-12 2011-06-30 Syntaxin Limited Suppression of cancers
AU2009259034B2 (en) 2008-06-12 2013-10-31 Ipsen Bioinnovation Limited Suppression of cancers
EP3590956A1 (en) 2008-06-12 2020-01-08 Ipsen Bioinnovation Limited Suppression of neuroendocrine diseases
MX2010014078A (en) 2008-06-25 2011-04-11 Braasch Biotech Llc Compositions and methods for enhanced somatostatin immunogenicity.
EP2356139A4 (en) 2008-07-23 2013-01-09 Harvard College Ligation of stapled polypeptides
GB0813873D0 (en) 2008-07-30 2008-09-03 Univ Dundee Compounds
WO2010028288A2 (en) 2008-09-05 2010-03-11 Aueon, Inc. Methods for stratifying and annotating cancer drug treatment options
EP2334317B1 (en) 2008-09-16 2017-06-14 The Research Foundation Of State University Of New York Stapled peptides and method of synthesis
CN102216322A (en) 2008-09-18 2011-10-12 纽约大学 Inhibiting interaction between the hif-1alpha and p300/cbp with hydrogen bond surrogate-based
US20110166063A1 (en) 2008-09-19 2011-07-07 Nektar Therapeutics Polymer conjugates of therapeutic peptides
JP2012503024A (en) 2008-09-22 2012-02-02 エルロン・セラピューティクス・インコーポレイテッド Peptidomimetic macrocycle
CN102884074A (en) 2008-09-22 2013-01-16 爱勒让治疗公司 Peptidomimetic macrocycles
AU2009294877C1 (en) 2008-09-22 2015-05-07 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
AU2009294869A1 (en) 2008-09-22 2010-03-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2010034029A1 (en) 2008-09-22 2010-03-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
JP2012503025A (en) 2008-09-22 2012-02-02 エルロン・セラピューティクス・インコーポレイテッド Method for preparing a purified polypeptide composition
EP2338056B1 (en) 2008-10-10 2018-01-03 Dana Farber Cancer Institute Chemical modulators of pro-apoptotic bax and bcl-2 polypeptides
JP2010120881A (en) 2008-11-19 2010-06-03 Keio Gijuku PEPTIDE FOR INHIBITING INTERACTION BETWEEN HUMAN CANCER PROTEIN MDM2 AND HUMAN CANCER-INHIBITING PROTEIN p53, AND USE THEREOF
KR101298168B1 (en) 2008-11-21 2013-08-20 충남대학교산학협력단 Chemical inhibitor of snail―p53 binding and pharmaceutical composition for treating cancer disease comprising the same
EP2352507A4 (en) 2008-11-24 2012-04-25 Aileron Therapeutics Inc Peptidomimetic macrocycles with improved properties
WO2010065572A1 (en) 2008-12-04 2010-06-10 The Salk Institute For Biological Studies Sstr1-selective analogs
EP3549951A3 (en) 2008-12-09 2019-11-20 Dana Farber Cancer Institute, Inc. Methods and compositions for specific modulation of mcl-1
US20100152114A1 (en) 2008-12-12 2010-06-17 Univ Of Miami And Usa By Dept Of Veterans Affairs Antioxidant activity of GH-RH Antagonists
BRPI1006139A2 (en) 2009-01-14 2017-05-30 Aileron Therapeutics Inc peptidomimetic macrocycles
WO2010083501A2 (en) 2009-01-16 2010-07-22 University Of South Florida Alpha-helix mimetic using a 2,5-oligopyrimidine scaffold
US8217051B2 (en) 2009-02-17 2012-07-10 Hoffmann-La Roche Inc. Spiroindolinone derivatives
US20100239589A1 (en) 2009-02-23 2010-09-23 Salk Institute For Biological Studies Methods and Compositions for Ameliorating Diabetes and Symptoms Thereof
FR2942798B1 (en) 2009-03-05 2011-04-08 Centre Nat Rech Scient PEPTIDES USEFUL FOR THE TREATMENT OF CHRONIC LYMPHOCYTIC LEUKEMIA
US20120135089A1 (en) 2009-03-17 2012-05-31 Stockwell Brent R E3 ligase inhibitors
US20100267636A1 (en) 2009-04-20 2010-10-21 Theratechnologies Inc. Use of cytochrome p450-metabolized drugs and grf molecules in combination therapy
US8076482B2 (en) 2009-04-23 2011-12-13 Hoffmann-La Roche Inc. 3,3′-spiroindolinone derivatives
US8883721B2 (en) 2009-05-12 2014-11-11 Mcgill University Methods of inhibiting the ghrelin/growth hormone secretatogue receptor pathway and uses thereof
CN101565390A (en) * 2009-05-27 2009-10-28 北京欧凯纳斯科技有限公司 Method for preparing Alpha-methyl-Alpha, Alpha-disubstituted-Alpha-aminophenol and derivatives thereof
US20100303794A1 (en) 2009-05-29 2010-12-02 Allergan, Inc. Methods of Treating Urogenital-Neurological Disorders Using Glucagon Like Hormone Retargeted Endopepidases
US20100303791A1 (en) 2009-05-29 2010-12-02 Allergan, Inc. Methods of Treating Chronic Neurogenic Inflammation Using Glucagon Like Hormone Retargeted Endopepidases
WO2011005219A1 (en) 2009-07-07 2011-01-13 Agency For Science, Technology And Research Novel mdm2 binding peptides and uses thereof
US9163330B2 (en) 2009-07-13 2015-10-20 President And Fellows Of Harvard College Bifunctional stapled polypeptides and uses thereof
BR112012008075A2 (en) 2009-08-26 2016-03-01 Novartis Ag tetrasubstituted heteroaryl compounds and their use as mdm2 and / or mdm4 modulators
CA2774973A1 (en) 2009-09-22 2011-03-31 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
IN2012DN03297A (en) 2009-09-30 2015-10-23 Tranzyme Pharma Inc
AU2010306718A1 (en) 2009-10-14 2012-05-24 Aileron Therapeutics, Inc. Improved peptidomimetic macrocycles
US8017607B2 (en) 2009-10-14 2011-09-13 Hoffmann-La Roche Inc. N-substituted-pyrrolidines as inhibitors of MDM2-P-53 interactions
WO2011053821A1 (en) 2009-10-30 2011-05-05 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
WO2011056961A2 (en) 2009-11-04 2011-05-12 Health Research Inc. Method and compositions for suppression of aging
PE20121282A1 (en) 2009-11-12 2012-10-12 Univ Michigan SPIRO-OXINDOL ANTAGONISTS OF MDM2
US20110118283A1 (en) 2009-11-17 2011-05-19 Qingjie Ding Substituted Pyrrolidine-2-Carboxamides
US8088815B2 (en) 2009-12-02 2012-01-03 Hoffman-La Roche Inc. Spiroindolinone pyrrolidines
CU24130B1 (en) 2009-12-22 2015-09-29 Novartis Ag ISOQUINOLINONES AND REPLACED QUINAZOLINONES
US8440693B2 (en) 2009-12-22 2013-05-14 Novartis Ag Substituted isoquinolinones and quinazolinones
US8658170B2 (en) 2010-01-06 2014-02-25 Joseph P. Errico Combination therapy with MDM2 and EFGR inhibitors
KR101220516B1 (en) 2010-01-21 2013-01-10 연세대학교 산학협력단 Human Adult Stem Cells Secreting Anti-MDM2 and Uses thereof
WO2011097642A1 (en) 2010-02-08 2011-08-11 Trustees Of Boston College Efficient methods for z- or cis-selective cross-metathesis
US8288431B2 (en) 2010-02-17 2012-10-16 Hoffmann-La Roche Inc. Substituted spiroindolinones
WO2011106650A2 (en) 2010-02-27 2011-09-01 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Novel p53-mdm2/p53-mdm4 antagonists to treat proliferative disease
WO2011127058A2 (en) 2010-04-09 2011-10-13 The Regents Of The University Of Michigan Biomarkers for mdm2 inhibitors for use in treating disease
WO2011133948A2 (en) 2010-04-22 2011-10-27 Longevity Biotech, Inc. Highly active polypeptides and methods of making and using the same
CN103179976A (en) 2010-05-13 2013-06-26 印第安纳大学研究及科技有限公司 Glucagon superfamily peptides exhibiting g protein-coupled receptor activity
WO2011143209A1 (en) 2010-05-13 2011-11-17 Indiana University Research And Technology Corporation Glucagon superfamily peptides exhibiting nuclear hormone receptor activity
US8980249B2 (en) 2010-06-03 2015-03-17 University Of Miami Agonists of growth hormone releasing hormone as effectors for survival and proliferation of pancreatic islets
CA2805406A1 (en) 2010-06-16 2011-12-22 Mcgill University Growth hormone secretatogue receptor antagonists and uses thereof
US20110313167A1 (en) 2010-06-22 2011-12-22 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Substituted Heterocycles as Therapeutic agents for treating cancer
US20130177979A1 (en) 2010-06-22 2013-07-11 University Of Central Florida Research Foundation, Inc. Methods and compositions for cell permeable stat3 inhibitor
MX2012014573A (en) 2010-06-24 2013-02-21 Univ Indiana Res & Tech Corp Dipeptide linked medicinal agents.
CN103179979A (en) 2010-06-24 2013-06-26 印第安纳大学研究及科技有限公司 Amide based glucagon superfamily peptide prodrugs
US8907053B2 (en) 2010-06-25 2014-12-09 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
JP2013532172A (en) * 2010-07-09 2013-08-15 ダナ ファーバー キャンサー インスティテュート インコーポレイテッド Stabilized insulin secretagogue peptides and methods of use
WO2012012352A2 (en) 2010-07-19 2012-01-26 Amidebio, Llc Modified peptides and proteins
US8975232B2 (en) 2010-07-29 2015-03-10 President And Fellows Of Harvard College Macrocyclic kinase inhibitors and uses thereof
DK2603600T3 (en) 2010-08-13 2019-03-04 Aileron Therapeutics Inc PEPTIDOMIMETIC MACROCYCLES
EP2614051A4 (en) 2010-09-08 2014-01-29 Univ Pittsburgh P53-mdm2 antagonists
US20120065210A1 (en) 2010-09-15 2012-03-15 Xin-Jie Chu Substituted hexahydropyrrolo[1,2-c]imidazolones
EP2431035A1 (en) 2010-09-16 2012-03-21 Æterna Zentaris GmbH Novel Triazole Derivatives with Improved Receptor Activity and Bioavailability Properties as Ghrelin Antagonists of Growth Hormone Secretagogue Receptors
US20130261058A1 (en) 2010-09-16 2013-10-03 University Of Miami Acceleration of wound healing by growth hormone releasing hormone and its agonists
CN102399283B (en) 2010-09-17 2013-05-29 中国农业大学 Mink growth hormone releasing hormone cDNA and application thereof
CN102399284B (en) 2010-09-17 2013-05-29 中国农业大学 Fox growth hormone releasing hormone cDNA and its application
US20120071499A1 (en) 2010-09-20 2012-03-22 Xin-Jie Chu Substituted Spiro[3H-Indole-3,6'(5'H)-[1H]Pyrrolo[1,2c]Imidazole-1',2(1H,2'H)-diones
WO2012040459A2 (en) 2010-09-22 2012-03-29 President And Fellows Of Harvard College Beta-catenin targeting peptides and uses thereof
US20130225603A1 (en) 2010-09-27 2013-08-29 Serrata Llc Mdm2 inhibitors for treatment of ocular conditions
EP2621275B1 (en) 2010-09-30 2018-03-28 St. Jude Children's Research Hospital Aryl-substituted imidazoles
EP2627662B1 (en) 2010-10-13 2015-09-16 Bristol-Myers Squibb Company Methods for preparing macrocycles and macrocycle stabilized peptides
FR2967072B1 (en) 2010-11-05 2013-03-29 Univ Dundee PROCESS FOR IMPROVING INFLUENZA PRODUCTION OF VACCINE AND SEEDS
CA2817568A1 (en) 2010-11-12 2012-05-18 The Salk Institute For Biological Studies Intellectual Property And Tech Nology Transfer Cancer therapies and diagnostics
UA109464C2 (en) 2010-11-12 2015-08-25 Дзе Ріджентс Оф Дзе Юніверсіті Оф Мічіган Spiro-oxindoles as mdm2 antagonists
WO2012076513A1 (en) 2010-12-09 2012-06-14 F. Hoffmann-La Roche Ag 3-cyano-1-hydroxymethyl-2-phenylpyrrolidine derivatives as inhibitors of mdm2-p53 interactions useful for the treatment of cancer
US9029332B2 (en) 2010-12-15 2015-05-12 The Research Foundation For The State University Of New York Cross-linked peptides and proteins, methods of making same, and uses thereof
RU2013131444A (en) 2010-12-16 2015-01-27 Рош Гликарт Аг COMBINED THERAPY BY AFUCOSYLED ANTIBODY TO CD20 AND MDM2 INHIBITOR
WO2012083181A1 (en) 2010-12-16 2012-06-21 Indiana University Research And Technology Corporation Alpha helix mimetics and methods for using
AR084308A1 (en) 2010-12-17 2013-05-08 Syngenta Participations Ag INSECTICIDE COMPOUNDS DERIVED FROM TRIAZOL
EP2474625B1 (en) 2011-01-05 2016-11-02 Daniela Kandioler Method for determining the p53 status of a tumour
EP2474624B1 (en) 2011-01-05 2016-08-17 Daniela Kandioler Response prediction in cancer treatment (p53 adapted cancer therapy)
WO2012094755A1 (en) 2011-01-14 2012-07-19 Theratechnologies Inc. Assessment of igf-1 levels in hiv-infected subjects and uses thereof
JP5950587B2 (en) 2011-02-28 2016-07-13 キヤノン株式会社 Method for producing porous glass and method for producing optical member
WO2012122059A1 (en) 2011-03-04 2012-09-13 New York University Hydrogen bond surrogate macrocycles as modulators of ras
CN103415530A (en) 2011-03-09 2013-11-27 Jitsubo株式会社 Novel cross-linked peptides containing non-peptide cross-linked structure, method for synthesizing cross-linked peptides, and novel organic compound used in method
TWI494312B (en) 2011-03-10 2015-08-01 Daiichi Sankyo Co Ltd Dispiropyrrolidine derivatives
WO2012149563A1 (en) 2011-04-29 2012-11-01 Kinemed, Inc. Chemical modification of apolipoprotein mimetic peptides for the production of therapeutic agents
WO2012173846A2 (en) 2011-06-06 2012-12-20 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20140256912A1 (en) 2011-06-17 2014-09-11 President And Fellows Of Harvard College Stabilized Variant MAML Peptides and Uses Thereof
WO2012174423A1 (en) 2011-06-17 2012-12-20 President And Fellows Of Harvard College Stabilized polypeptides as regulators of rab gtpase function
GB201110390D0 (en) 2011-06-20 2011-08-03 Medical Res Council Compounds for use in stabilising p53 mutants
US20120328692A1 (en) 2011-06-24 2012-12-27 University Of Maryland, Baltimore Potent d-peptide antagonists of mdm2 and mdmx for anticancer therapy
EP3041491A1 (en) 2011-08-31 2016-07-13 New York University Thioether-,ether-, and alkylamine-linked hydrogen bond surrogate pertidomimentics
EP3444262A3 (en) 2011-09-09 2019-04-10 Agency For Science, Technology And Research P53 activating peptides
WO2013049250A1 (en) 2011-09-27 2013-04-04 Amgen Inc. Heterocyclic compounds as mdm2 inhibitors for the treatment of cancer
US9096684B2 (en) 2011-10-18 2015-08-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2013059530A2 (en) 2011-10-18 2013-04-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9408885B2 (en) 2011-12-01 2016-08-09 Vib Vzw Combinations of therapeutic agents for treating melanoma
CA2761253A1 (en) 2011-12-07 2013-06-07 Vib Vzw Combinations of therapeutic agents for treating melanoma
AU2013221432B2 (en) 2012-02-15 2018-01-18 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2013123267A1 (en) 2012-02-15 2013-08-22 Aileron Therapeutics, Inc. Triazole-crosslinked and thioether-crosslinked peptidomimetic macrocycles
US9890429B2 (en) 2012-02-29 2018-02-13 Dana-Farber Cancer Institute, Inc. Compositions, kits, and methods for the identification, assessment, prevention, and therapy of cancer
WO2013131019A1 (en) 2012-03-02 2013-09-06 Ludwig Institute For Cancer Research Ltd. Iaspp phosphorylation and metastatic potential
US9216170B2 (en) 2012-03-19 2015-12-22 Hoffmann-La Roche Inc. Combination therapy for proliferative disorders
WO2013166319A1 (en) 2012-05-02 2013-11-07 Kansas State University Research Foundation Macrocyclic and peptidomimetic compounds as broad-spectrum antivirals against 3c or 3c-like proteases of picornaviruses, caliciviruses and coronaviruses
US20150159222A1 (en) 2012-07-31 2015-06-11 Novartis Ag Markers associated with human double minute 2 inhibitors
CA3113959A1 (en) 2012-09-26 2014-04-03 President And Fellows Of Harvard College Proline-locked stapled peptides and uses thereof
US20150225471A1 (en) 2012-10-01 2015-08-13 President And Fellows Of Harvard College Stabilized polypeptide insulin receptor modulators
MX2015005244A (en) 2012-11-01 2015-07-14 Aileron Therapeutics Inc Disubstituted amino acids and methods of preparation and use thereof.
WO2014138429A2 (en) 2013-03-06 2014-09-12 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and use thereof in regulating hif1alpha
EP2991665A4 (en) 2013-03-13 2016-11-09 Harvard College Stapled and stitched polypeptides and uses thereof
US20140288116A1 (en) 2013-03-15 2014-09-25 Life Technologies Corporation Classification and Actionability Indices for Lung Cancer
US9198910B2 (en) 2013-04-04 2015-12-01 The Translational Genomics Research Institute Methods for the treatment of cancer
ES2782003T3 (en) 2013-04-16 2020-09-09 Memorial Sloan Kettering Cancer Center Diagnostic Companion for CDK4 Inhibitors
CA2912547A1 (en) 2013-07-03 2015-01-08 F. Hoffmann-La Roche Ag Mrna-based gene expression for personalizing patient cancer therapy with an mdm2 antagonist
US9268662B2 (en) 2013-08-01 2016-02-23 Oracle International Corporation Method and system for a high availability framework
WO2015017803A1 (en) 2013-08-02 2015-02-05 Children's Hospital Medical Center Compositions and methods for treating disease states associated with activated t cells and/or b cells
US20160115556A1 (en) 2013-10-19 2016-04-28 Trovagene, Inc. Detecting mutations in disease over time
KR20220167335A (en) 2013-11-11 2022-12-20 암젠 인크 Combination therapy including an mdm2 inhibitor and one or more additional pharmaceutically active agents for the treatment of cancers
MX370618B (en) 2013-12-05 2019-12-17 Hoffmann La Roche Novel combination treatment for acute myeloid leukemia (aml).
MX2016008362A (en) 2013-12-23 2016-09-08 Novartis Ag Pharmaceutical combinations.
US20160333419A1 (en) 2014-01-14 2016-11-17 Daiichi Sankyo Company, Limited Gene signatures associated with sensitivity to mdm2 inhibitors
WO2015157508A1 (en) 2014-04-09 2015-10-15 Aileron Therapeutics, Inc. Peptidomimetic macrocycles with pth activity
WO2015179799A1 (en) 2014-05-22 2015-11-26 The General Hospital Corporation DD1alpha RECEPTOR AND USES THEREOF IN IMMUNE DISORDERS
TW201613576A (en) 2014-06-26 2016-04-16 Novartis Ag Intermittent dosing of MDM2 inhibitor
MX2017003227A (en) 2014-09-13 2017-12-04 Novartis Ag Combination therapies of alk inhibitors.
WO2016049359A1 (en) 2014-09-24 2016-03-31 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
MX2017003819A (en) 2014-09-24 2017-06-15 Aileron Therapeutics Inc Peptidomimetic macrocycles and formulations thereof.
EP3204514A1 (en) 2014-10-09 2017-08-16 Daiichi Sankyo Co., Ltd. Algorithms for gene signature-based predictor of sensitivity to mdm2 inhibitors
JP6608439B2 (en) 2014-10-10 2019-11-20 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト Methods for personalizing cancer treatment of patients with MDM2 antagonists
WO2016073184A1 (en) 2014-11-04 2016-05-12 Dana Farber Cancer Institute, Inc. Compositions and methods for treating multiple myeloma
CN107223163A (en) 2014-12-24 2017-09-29 豪夫迈·罗氏有限公司 For the treatment of bladder cancer, diagnosis and method of prognosis
WO2016154058A1 (en) 2015-03-20 2016-09-29 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10059741B2 (en) 2015-07-01 2018-08-28 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
AU2016287754B2 (en) 2015-07-02 2021-02-25 Dana-Farber Cancer Institute, Inc. Stabilized anti-microbial peptides
WO2017023933A2 (en) 2015-08-03 2017-02-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
CN108348571B (en) 2015-09-03 2022-03-22 艾瑞朗医疗公司 Peptidomimetic macrocycles and uses thereof
CN108368161A (en) 2015-09-10 2018-08-03 艾瑞朗医疗公司 Peptidomimetic macrocyclic compound as MCL-1 conditioning agents
JP2019520304A (en) 2016-03-21 2019-07-18 エルロン・セラピューティクス・インコーポレイテッドAileron Therapeutics,Inc. Companion diagnostic tool for peptidomimetic macrocycles
WO2017205786A1 (en) 2016-05-27 2017-11-30 Aileron Therapeutics, Inc. Cell permeable peptidomimetic macrocycles
WO2017218949A2 (en) 2016-06-17 2017-12-21 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US20190185518A9 (en) 2017-03-09 2019-06-20 Aileron Therapeutics, Inc. Warhead-containing peptidomimetic macrocycles as modulators of bfl-1

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7786072B2 (en) * 1999-05-18 2010-08-31 President And Fellows Of Harvard College Stabilized compounds having secondary structure motifs

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
"Purification of Laboratory Chemicals" fifth edition, 2003, Armarego et al. *

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9175056B2 (en) 2006-12-14 2015-11-03 Alleron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
US10328117B2 (en) 2006-12-14 2019-06-25 Aileron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
US9675661B2 (en) 2006-12-14 2017-06-13 Aileron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
US9493509B2 (en) 2007-02-23 2016-11-15 Aileron Therapeutics, Inc. Triazole macrocycle systems
US9957296B2 (en) 2007-02-23 2018-05-01 Aileron Therapeutics, Inc. Triazole macrocycle systems
US10030049B2 (en) 2007-02-23 2018-07-24 Aileron Therapeutics, Inc. Triazole macrocycle systems
US10301351B2 (en) 2007-03-28 2019-05-28 President And Fellows Of Harvard College Stitched polypeptides
US9175045B2 (en) 2008-09-22 2015-11-03 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9458202B2 (en) 2008-11-24 2016-10-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles with improved properties
US10022422B2 (en) 2009-01-14 2018-07-17 Alleron Therapeutics, Inc. Peptidomimetic macrocycles
US10300109B2 (en) 2009-09-22 2019-05-28 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9957299B2 (en) 2010-08-13 2018-05-01 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10308699B2 (en) 2011-10-18 2019-06-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9522947B2 (en) 2011-10-18 2016-12-20 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9096684B2 (en) 2011-10-18 2015-08-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10227380B2 (en) 2012-02-15 2019-03-12 Aileron Therapeutics, Inc. Triazole-crosslinked and thioether-crosslinked peptidomimetic macrocycles
US10213477B2 (en) 2012-02-15 2019-02-26 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9505804B2 (en) 2012-02-15 2016-11-29 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9845287B2 (en) 2012-11-01 2017-12-19 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US10669230B2 (en) 2012-11-01 2020-06-02 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US10987427B2 (en) 2013-09-13 2021-04-27 The Scripps Research Institute Modified therapeutic agents and compositions thereof
US10039809B2 (en) 2013-12-18 2018-08-07 The California Institute For Biomedical Research Modified therapeutic agents, stapled peptide lipid conjugates, and compositions thereof
US11007252B2 (en) 2013-12-18 2021-05-18 The Scripps Research Institute Modified therapeutic agents, stapled peptide lipid conjugates, and compositions thereof
US11865160B2 (en) 2013-12-18 2024-01-09 The Scripps Research Institute Modified therapeutic agents, stapled peptide lipid conjugates, and compositions thereof
US10471120B2 (en) 2014-09-24 2019-11-12 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10905739B2 (en) 2014-09-24 2021-02-02 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and formulations thereof
US10253067B2 (en) 2015-03-20 2019-04-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10059741B2 (en) 2015-07-01 2018-08-28 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10023613B2 (en) 2015-09-10 2018-07-17 Aileron Therapeutics, Inc. Peptidomimetic macrocycles as modulators of MCL-1

Also Published As

Publication number Publication date
US9604919B2 (en) 2017-03-28
US20170066714A1 (en) 2017-03-09
EP2914256A1 (en) 2015-09-09
IL238084A0 (en) 2015-05-31
AU2013337388B2 (en) 2018-08-02
CA2887285A1 (en) 2014-05-08
US9845287B2 (en) 2017-12-19
JP2016503404A (en) 2016-02-04
US10669230B2 (en) 2020-06-02
EP2914256A4 (en) 2016-06-22
KR20150082307A (en) 2015-07-15
MX2015005244A (en) 2015-07-14
ZA201502991B (en) 2016-01-27
CN104812384B (en) 2020-09-18
AU2013337388A1 (en) 2015-04-30
SG11201503052RA (en) 2015-05-28
WO2014071241A1 (en) 2014-05-08
JP2019059777A (en) 2019-04-18
EP2914256B1 (en) 2019-07-31
JP6526563B2 (en) 2019-06-05
BR112015009470A2 (en) 2019-12-17
US20180265459A1 (en) 2018-09-20
CN104812384A (en) 2015-07-29

Similar Documents

Publication Publication Date Title
US10669230B2 (en) Disubstituted amino acids and methods of preparation and use thereof
US20230406879A1 (en) Method for synthesizing peptide containing n-substituted amino acid
JP6350632B2 (en) Method for producing peptide
JP6011528B2 (en) Method for producing peptide
JP2002525376A (en) Auxiliary groups for amide bond formation
US9850285B2 (en) Process for preparing eptifibatide
EP4251632A1 (en) Compositions and methods for chemical synthesis
JP6788376B2 (en) Method for Producing Peptide Containing Aspartic Acid Residue
US9605020B2 (en) Method for producing dipeptide derivative containing disubstituted amino acid residue
CA2779949A1 (en) Peptidomimetics comprising n-amino cyclic urea residues and uses thereof
US20230037643A1 (en) Method for producing peptide
WO2013024720A1 (en) PEPTIDE CATALYST AND ALDEHYDE ASYMMETRIC α-AMINATION USING SAME

Legal Events

Date Code Title Description
AS Assignment

Owner name: AILERON THERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DARLAK, KRZYSZTOF;KAWAHATA, NORIYUKI;ATHAMNEH, SAMEER AHMED;SIGNING DATES FROM 20140919 TO 20141008;REEL/FRAME:035511/0786

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4